Download as pdf or txt
Download as pdf or txt
You are on page 1of 554

Tumor Ablation

Tumor Ablation
Principles and Practice

Eric vanSonnenberg, MD
William N. McMullen
Luigi Solbiati, MD
Editors

Tito Livraghi, MD
Peter R. Müeller, MD
Stuart G. Silverman, MD

Associate Editors

With 200 Illustrations in 457 Parts


Eric vanSonnenberg, MD William N. McMullen
formerly: Interventional Oncology Consultant
Chief of Radiology Boston, MA 02115
Dana-Farber Cancer Institute USA
Harvard Medical School
Interventional Radiologist
Brigham and Women’s Hospital
Children’s Hospital
Boston, MA 02115
USA
Luigi Solbiati, MD
Chairman
Department of Radiology
Ospedale Civile Busto Arsizio
21052 Busto Arsizio
Italy

Library of Congress Cataloging-in-Publication Data


Tumor ablation: principles and practice / [edited by] Eric vanSonnenberg, William
McMullen, Luigi Solbiati.
p. ; cm.
Includes bibliographical references and index.
ISBN 0-387-95539-9 (alk. paper)
1. Cancer—Thermotherapy. 2. Catheter ablation. I. vanSonnenberg, Eric.
II. McMullen, William. III. Solbiati, L. (Luigi)
[DNLM: 1. Neoplasms—therapy. 2. Catheter Ablation—methods. QZ 266 T9232
2004]
RC271.T5T865 2004
616.99¢40632—dc22 2004049184

ISBN 10: 0-387-95539-9 Printed on acid-free paper.


ISBN 13: 978-0387-95539-1

© 2005 Springer Science+Business Media, Inc.

All rights reserved. This work may not be translated or copied in whole or in part without
the written permission of the publisher (Springer Science+Business Media, Inc., 233 Spring
Street, New York, NY 10013, USA), except for brief excerpts in connection with reviews
or scholarly analysis. Use in connection with any form of information storage and retrieval,
electronic adaptation, computer software, or by similar or dissimilar methodology now
known or hereafter developed is forbidden.
The use in this publication of trade names, trademarks, service marks, and similar terms,
even if they are not identified as such, is not to be taken as an expression of opinion as to
whether or not they are subject to proprietary rights.
While the advice and information in this book are believed to be true and accurate at the
date of going to press, neither the authors nor the editors nor the publisher can accept any
legal responsibility for any errors or omissions that may be made. The publisher makes no
warranty, express or implied, with respect to the material contained herein.

Printed in the United States of America. (BS/MVY)

9 8 7 6 5 4 3 2 1 SPIN 10885664

springeronline.com
Foreword I

There is an enormous sense of excitement in the communities of cancer


research and cancer care as we move into the middle third of the first
decade of the 21st century. For the first time, there is a true sense of con-
fidence that the tools provided by the human genome project will enable
cancer researchers to crack the code of genomic abnormalities that allow
tumor cells to live within the body and provide highly specific, virtually
non-toxic therapies for the eradication, or at least firm control of human
cancers. There is also good reason to hope that these same lines of
inquiry will yield better tests for screening, early detection, and preven-
tion of progression beyond curability.
While these developments provide a legitimate basis for much opti-
mism, many patients will continue to develop cancers and suffer from
their debilitating effects, even as research moves ahead. For these indi-
viduals, it is imperative that the cancer field make the best possible use
of the tools available to provide present day cancer patients with the best
chances for cure, effective palliation, or, at the very least, relief from
symptoms caused by acute intercurrent complications of cancer. A
modality that has emerged as a very useful approach to at least some of
these goals is tumor ablation by the use of physical or physiochemical
approaches. Tumor ablation by means of other than traditional surgical
approaches, as noted in the excellent Introduction to this text, is a
methodology attempted with modest success for a number of years.
Modern approaches take advantage of the vastly superior armamen-
tarium of imaging strategies now available. These permit pinpoint local-
ization of tumors causing distressing symptoms, such as pain, obstruction,
compression, etc. Advances in materials science, and methods for deliv-
ery of ablating agents such as intense cold, ultrasound, etc., combined
with improved localization now make it possible to be much more
aggressive and effective in attempting to achieve local ablation of
primary or metastatic tumors that cause morbidity.
This book represents an outstanding effort on the part of the editors
and authors to gather in one place a review of the advances in tumor
ablation methods as well as a clear, practical, and readable description
of specific applications in different organ systems. The sections on
imaging of tumors for the purposes of localizing targets for the ablation

v
vi Foreword I

methodologies that are covered in the next section, provide physicians


involved in cancer research, regardless of orientation, with an indis-
pensable resource for understanding the rationale, capabilities, limita-
tions, benefits, and toxicities of these approaches. Readers of this volume
will no doubt find a strong basis for an increased, but much more appro-
priate, use of this method for the benefit of their patients with cancers
that are either advanced or located in spots difficult to reach by surgery,
radiation therapy, or chemotherapeutic drugs. In this regard, the final
section offers perspectives from cancer care experts who approach the
care of these patients from a variety of perspectives: the medical oncol-
ogist’s, the surgical oncologist’s, the radiation therapist’s, etc. are espe-
cially valuable. It provides a good balance between those who are
immersed in developing and advancing this modality and the “end users,”
who must balance the use of these methods with a variety of other
complex options. The final section of this text is also valuable, because it
provides a basis for approaching the difficult issue of how to integrate a
novel modality of therapy into the totality of cancer care to the benefit
of the individual patients.
In my view, this book provides a comprehensive yet incisive overview
of a field that has reached a highly appropriate stage for this kind of
resource. Tumor ablation, at least in its present form, is still a relatively
young field that can be adequately captured in a volume of this size. On
the other hand, it has grown sufficiently large in its methods and appli-
cations that a summary is needed both within and outside the field. Dr.
vanSonnenberg is to be complimented for taking on this difficult chal-
lenge and meeting it so effectively.

Edward J. Benz, Jr, MD


President of Dana-Farber
Cancer Institute
Professor of Medical Oncology
& Hematology
Harvard Medical School
Foreword II

Drs. vanSonnenberg, McMullen, and Solbiati have produced a truly


timely and comprehensive textbook on a topic that is coming of age.
While the last few decades have seen the development and establish-
ment of safe surgery for malignant tumors, forward thinking surgeons,
radiologists, and interventional radiologists have now begun to consider
and realize the potential to treat solid tumors with ablative techniques.
Although not a new concept, the use of extreme temperatures to kill
tumors but preserve normal cells has exploded in recent years. As with
any new treatment modality, more questions are often raised than
answers. This textbook provides the basis from which one can draw infor-
mation on virtually any topic related to ablative procedures for tumors,
and should be an essential resource for anyone who treats solid tumors
potentially amenable to ablation.
This textbook is not only timely, but it is comprehensive. It addresses
all aspects of ablation through contributions from the leading experts in
the field. The spectrum of topics addressed by the editors is appropriate
and is exemplary for its implicit understanding that this is a multi-
disciplinary field that requires the teamwork of specialists in many fields
for the ultimate success of ablative techniques. From basic science, to the
logistics of practice, to imaging, to specific tumors, and to psychosocial
issues, this book has tremendous breadth of coverage, giving it extraor-
dinary value to students and practitioners. As we embark on more and
more innovative approaches to the treatment of solid tumors, textbooks
such as this are an essential reference for anyone interested in practic-
ing or researching ablative techniques.

Leslie H. Blumgart, MD
Michael I. D’Angelica, MD
Department of Surgery
Memorial Sloan-Kettering Hospital
Cornell Medical School

vii
Foreword III

Radiofrequency ablation (thermal tissue coagulation) has evolved


rapidly in the decade since I was first enticed to use two simple bipolar
needle electrodes in the laboratory in an attempt to produce coagulative
necrosis of liver tissue. Important preclinical and clinical work from the
authors included in this book paved the way for the clinical trials utiliz-
ing radiofrequency ablation as a treatment for unresectable primary and
secondary hepatic malignancies. As I predicted to many of the engineers
in the industry involved with radiofrequency ablation equipment, “build
it and they will come”; investigators have subsequently employed local
thermal destruction of tumors in many additional body sites and organs.
Several chapters in this book describe the current status of radio-
frequency ablation of tumors in organs other than the liver.
Technologic advances have produced an ever-growing array of multi-
ple needle and hook electrode arrays. Ongoing research in industry and
academic centers is designed to produce larger zones of thermal necro-
sis in a safe and reproducible fashion. The geometry of zones of thermal
necrosis produced by radiofrequency is a complex issue, and numerous
mathematical models have been developed in an attempt to understand
the overlap of zones of thermal necrosis necessary to treat larger hepatic
malignancies. Unfortunately, many of these models fail to account for the
effect of large blood vessels near the treated tumors or the differences
in the blood flow between tumor and normal tissue. Like many novel
treatments that involve technical skill and the manipulation of instru-
ments, there is a learning curve when utilizing radiofrequency ablation
to treat malignancies in any body site. Another important aspect of
radiofrequency ablation that is reviewed in this book is the role and lim-
itations of imaging modalities to monitor and predict the completeness
of tumor destruction during radiofrequency ablation, and the role and
limitations of diagnostic imaging modalities to diagnose local recurrence
(incomplete tumor destruction) following radiofrequency ablation.
The challenges in the next decade regarding radiofrequency ablation
include establishing long-term disease-free and overall survival rates
following radiofrequency ablation of primary and metastatic hepatic
malignancies, developing criteria to better define those patients who are
candidates for treatment with a percutaneous approach versus those who

ix
Foreword III x

are better treated with a laparoscopic or open surgical radiofrequency


ablation procedure, defining the role and the relative risks and benefits
in treating non hepatic malignancies in other organs and body sites, and
developing and completing clinical trials involving advances in radio-
frequency ablation equipment and combined modalities to improve the
reliability and reduce local recurrence rates after treatment. The role,
benefits, risks, and complications of radiofrequency ablation to treat
malignant tumors must be identified and defined. Radiofrequency as a
part of multimodality therapies, including surgical resection of large
liver tumors combined with radiofrequency ablation of additional small
lesions, and the use of neoadjuvant and adjuvant treatments with
radiofrequency ablation of hepatic malignancies are being explored.
Exciting results should be forthcoming in the next several years. The field
is truly “heating up,” as other forms of thermal ablation treatments have
been introduced into the market place; the role of these modalities also
will be studied in the coming decade. The rapid progress in radio-
frequency ablation of hepatic and other tumors indicates that a signifi-
cant patient population is in need of a safe and reliable treatment to
produce complete destruction of measurable tumors.

Steven A. Curley, MD
Department of Surgery
M. D. Anderson Hospital
University of Texas
Preface

So much good is happening on so many fronts in the battle against


cancer—from powerful new drugs to complex anti-angiogenesis strate-
gies to creative research unlocking the mysteries of molecular biology to
thermal and mechanical methods of tumor destruction. The specialty of
radiology is contributing in integral and multifaceted ways in this anti-
cancer crusade. Remarkable advances in the precision and speed of CT,
MRI diffusion and spectroscopy, metabolic assessment by PET, and
avant garde fusion imaging with PET/CT, as well as percutaneous
methods to kill tumors, all have a powerful impact in the fight against
cancer.
The time is both ripe and right for a book on Tumor Ablation. There
is a rapidly growing acceptance, if not excitement, that this method adds
a new dimension to the therapeutic oncologic armamentarium. While
some forms of percutaneous ablation have been in existence for more
than ten years, a certain maturity has been achieved, such that referrals
from oncologists, surgeons, and radiation therapists are now common—
not to mention patient self-referral. While research in the ablation field
expands and innovations are constantly made, there has been a leveling
off of the exponential growth and changes in the ablation field. Hence,
larger series are now being accumulated, the procedures are proliferat-
ing in many hospitals, and there is multidisciplinary acceptance of abla-
tion as a viable alternative for select patients with cancer. The book
focuses on tumor ablation, largely but not exclusively percutaneous, with
guidance and monitoring by a variety of imaging modalities.
Section I covers the background and foundations of tumor ablation.
The building of a practice, anesthesia, and the various instruments for
ablation are described in Section II. Section III discusses imaging for
ablation. Section IV deals with the various methods of ablation. Section
V reviews the clinical experience by leaders in the field about a variety
of tumors in different organ systems. Section VI offers perspectives by
nonradiology oncologic physicians, along with future directions in the
field. Tumor Ablation contains forewords by oncologists and surgeons
from three prestigious cancer centers with frank insights into the role of
tumor ablation in oncology. A chapter that depicts the cancer journey by
patients and their families provides unique perspectives.

xi
xii Preface

Our goal was to assemble the pioneers and world’s experts in the field
of tumor ablation to contribute to the book; we succeeded, but more
importantly, they succeeded in conveying their marvelous creativity and
contributions. Extensive experience is provided by numerous authors
from Europe and Asia, where much of the seminal work originated. The
many North American experts highlight their pushing of the envelope,
reflected in many of the major advances in the field of tumor ablation.
Contributors include radiologists, surgeons, oncologists, anesthesiolo-
gists, nonphysician scientists, and internists, thereby reflecting the multi-
disciplinary nature of tumor ablation.
Finally, our appreciation to those who facilitated this endeavor. First
to our publisher, Rob “Louie” Albano, for his wise counsel, and to his
associate, Michelle Schmitt-DeBonis. Our thanks to our local assistants
for their help, Kristen Rancourt and Sue Ellen Lynch. Our appreciation
to all our contributing authors for sharing their expertise, experience, and
enlightenment. Finally to our patients and families—it has been, and is,
a privilege to have offered and delivered hope and benefit to their care.

Eric vanSonnenberg, MD
William N. McMullen
Luigi Solbiati, MD
Contents

Foreword I by Edward J. Benz, Jr. . . . . . . . . . . . . . . . . . . . . . . . . v


Foreword II by Leslie H. Blumgart and
Michael I. D’Angelica . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii
Foreword III by Steven A. Curley . . . . . . . . . . . . . . . . . . . . . . . . ix
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xvii

Section I Introduction to Ablation

1 History of Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
John P. McGahan and Vanessa A. van Raalte

2 Epidemiology: How to Appraise the


Ablation Literature Critically . . . . . . . . . . . . . . . . . . . . . . . 17
Craig Earle

3 Image-Guided Tumor Ablation: Basic Science . . . . . . . . . . 23


Muneeb Ahmed and S. Nahum Goldberg

4 Tumor Angiogenesis: General Principles and


Therapeutic Approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
John V. Heymach and Judah Folkman

Section II Operations for Tumor Ablation

5 Image-Guided Tumor Ablation: How to Build


a Practice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
Gary Onik

6 Anesthesia for Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . . 64


John A. Fox and Alan M. Harvey

xiii
xiv Contents

7 Devices, Equipment, and Operation of


Ablation Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
Paul R. Morrison and Eric vanSonnenberg

Section III Imaging for Tumor Ablation

8 Intraoperative Ultrasound-Guided Procedures . . . . . . . . . 95


Robert A. Kane

9 Computed Tomography Imaging for Tumor Ablation . . . . 104


Thierry de Baère

10 Positron Emission Tomograpy Imaging for


Tumor Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Annick D. Van den Abbeele, David A. Israel,
Stanislav Lechpammer, and Ramsey D. Badawi

11 Ultrasound Imaging in Tumor Ablation . . . . . . . . . . . . . . . 135


Massimo Tonolini and Luigi Solbiati

12 Magnetic Resonance Imaging Guidance for


Tumor Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 148
Koenraad J. Mortele, Stuart G. Silverman, Vito Cantisani,
Kemal Tuncali, Sridhar Shankar, and Eric vanSonnenberg

13 Magnetic Resonance Imaging Guidance of Radiofrequency


Thermal Ablation for Cancer Treatment . . . . . . . . . . . . . . 167
Daniel T. Boll, Jonathan S. Lewin, Sherif G. Nour, and
Elmar M. Merkle

14 Image Guidance and Control of Thermal Ablation . . . . . . 182


Ferenc A. Jolesz

Section IV Methods of Ablation

15 Percutaneous Ethanol Injection Therapy . . . . . . . . . . . . . . 195


Tito Livraghi and Maria Franca Meloni

16 Radiofrequency Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . 205


Riccardo Lencioni and Laura Crocetti

17a Microwave Coagulation Therapy for Liver Tumors . . . . . . 218


Toshihito Seki

17b Microwave Ablation: Surgical Perspective . . . . . . . . . . . . . 228


Andrew D. Strickland, Fateh Ahmed, and David M. Lloyd
Contents xv

18 Percutaneous Laser Therapy of Primary and Secondary


Liver Tumors and Soft Tissue Lesions: Technical Concepts,
Limitations, Results, and Indications . . . . . . . . . . . . . . . . . 234
Thomas J. Vogl, Ralf Straub, Katrin Eichler, Stefan Zangos,
and Martin Mack

19 Cryoablation: History, Mechanism of Action, and


Guidance Modalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
Sharon M. Weber and Fred T. Lee, Jr.

20 Combined Regional Chemoembolization and Ablative


Therapy for Hepatic Malignancies . . . . . . . . . . . . . . . . . . . 266
Michael C. Soulen and Lily Y. Kernagis

21 Focused Ultrasound for Tumor Ablation . . . . . . . . . . . . . . 273


Clare Tempany, Nathan MacDonold,
Elizabeth A. Stewart, and Kullervo Hynynen

22 New Technologies in Tumor Ablation . . . . . . . . . . . . . . . . . 285


Bradford J. Wood, Ziv Neeman, and Anthony Kam

23 Combination Therapy for Ablation . . . . . . . . . . . . . . . . . . 301


Allison Gillams and William R. Lees

Section V Organ System Tumor Ablation

24 Ablation of Liver Metastases . . . . . . . . . . . . . . . . . . . . . . . 311


Luigi Solbiati, Tiziana Ierace, Massimo Tonolini, and
Luca Cova

25 Ablation for Hepatocellular Carcinoma . . . . . . . . . . . . . . . 322


Maria Franca Meloni and Tito Livraghi

26 Radiofrequency Ablation of
Neuroendocrine Metastases . . . . . . . . . . . . . . . . . . . . . . . . 332
Thomas D. Atwell, J. William Charboneau,
David M. Nagorney, and Florencia G. Que

27 Tumor Ablation in the Kidney . . . . . . . . . . . . . . . . . . . . . . 341


Debra A. Gervais and Peter R. Müeller

28 Radiofrequency Ablation for Thoracic Neoplasms . . . . . . . 353


Sapna K. Jain and Damian E. Dupuy

29 Soft Tissue Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 369


Sridhar Shankar, Eric vanSonnenberg, Stuart G. Silverman,
Paul R. Morrison, and Kemal Tuncali
xvi Contents

30 Image-Guided Palliation of Painful


Skeletal Metastases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 377
Matthew R. Callstrom, J. William Charboneau,
Matthew P. Goetz, and Joseph Rubin

31 Radiofrequency Ablation of Osteoid Osteoma . . . . . . . . . 389


Daniel I. Rosenthal and Hugue Ouellette

32 Image-Guided Prostate Cryotherapy . . . . . . . . . . . . . . . . . 402


Gary Onik

33 Uterine Artery Embolization for Fibroid Disease . . . . . . . 412


Robert L. Worthington-Kirsch

34 Applications of Cryoablation in the Breast . . . . . . . . . . . . 422


John C. Rewcastle

35 Percutaneous Ablation of Breast Tumors . . . . . . . . . . . . . . 428


Bruno D. Fornage and Beth S. Edeiken

36 Complications of Tumor Ablation . . . . . . . . . . . . . . . . . . . 440


Lawrence Cheung, Tito Livraghi, Luigi Solbiati,
Gerald D. Dodd III, and Eric vanSonnenberg

Section VI Perspectives

37 Tumor Ablation for Patients with Lung Cancer:


The Thoracic Oncologist’s Perspective . . . . . . . . . . . . . . . . 459
Bruce E. Johnson and Pasi A. Jänne

38 Ablative Therapies for Gastrointestinal Malignancies:


The Gastrointestinal Oncologist’s Viewpoint . . . . . . . . . . . 466
Matthew Kulke

39 Treatment of Hepatocellular Carcinoma by Internists . . . . 472


Shuichiro Shiina, Takuma Teratani, and Masao Omata

40 The Surgeon’s Perspective on Hepatic


Radiofrequency Ablation . . . . . . . . . . . . . . . . . . . . . . . . . . 480
David A. Iannitti

41 Radiofrequency Tumor Ablation in Children . . . . . . . . . . . 489


William E. Shiels II and Stephen D. Brown

42 Comments from Patients and Their Families . . . . . . . . . . . 498


Eric vanSonnenberg

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517
Contributors

Fateh Ahmed, MBBS


Surgical Research Fellow, Department of Surgery, Leicester Royal Infir-
mary, University Hospitals, Leicester, UK

Muneeb Ahmed, MD
Research Assistant in Radiology, Harvard Medical School, Beth Israel
Deaconess Medical Center, Boston, MA, USA

Thomas D. Atwell, MD
Senior Associate Consultant, Department of Radiology, Mayo Clinic
College of Medicine, Rochester, MN, USA

Ramsey D. Badawi, PhD


Assistant Professor, Department of Radiology, University of California
Davis, University of California Davis Medical Center, Sacramento, CA,
USA

Daniel T. Boll, MD
Research Associate, Department of Radiology, Case Western Reserve
University and University Hospitals of Cleveland, Cleveland, OH, USA

Stephen D. Brown, MD
Instructor in Radiology, Harvard Medical School, Staff Radiologist, Chil-
dren’s Hospital, Boston, MA, USA

Matthew R. Callstrom, MD, PhD


Assistant Professor of Radiology, Department of Radiology, Mayo Clinic
College of Medicine, Rochester, MN, USA

Vito Cantisani, MD
Department of Radiology, Brigham and Women’s Hospital, Boston, MA,
USA

J. William Charboneau, MD
Professor of Radiology, Department of Radiology, Mayo Clinic College
of Medicine, Rochester, MN, USA

xvii
xviii Contributors

Lawrence Cheung
Medical Student, Harvard Medical School, Boston, MA, USA

Luca Cova, MD
Department of Radiology, Ospedale Civile Busto Arsizio, Busto Arsizio,
Italy

Laura Crocetti, MD
Research Fellow, Department of Oncology, Transplants, and Advanced
Technologies in Medicine, University of Pisa, Pisa, Italy

Thierry de Baère, MD
Staff Radiologist, Institut Gustave Roussy, Villejuif, France

Gerald D. Dodd III, MD, FACR


Professor of Radiology, University of Texas, San Antonio, Chairman of
Radiology, University of Texas Health Science Center, San Antonio, TX,
USA

Damian E. Dupuy, MD
Professor of Diagnostic Imaging, Brown Medical School, Director of
Ultrasound and Office of Minimally Invasive Therapy, Rhode Island
Hospital, Providence, RI, USA

Craig Earle, MD, PhD


Assistant Professor of Medicine, Harvard Medical School, Department
of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA

Beth S. Edeiken, MD
Professor of Radiology, Department of Radiology, M. D. Anderson
Cancer Center, Houston, TX, USA

Katrin Eichler, MD
Institute for Diagnostic and Interventional Radiology, Department of
Radiology, University Hospital, Frankfurt, Germany

Judah Folkman, MD
Professor of Surgery, Harvard Medical School, Children’s Hospital,
Boston, MA, USA

Bruno D. Fornage, MD
Professor of Radiology, University of Texas, Houston, Department of
Radiology, M. D. Anderson Cancer Center, Houston, TX, USA

John A. Fox, MD
Staff Anesthesiologist, Department of Anesthesiology, Perioperative and
Pain Medicine, Brigham and Women’s Hospital, Boston, MA, USA

Debra A. Gervais, MD
Assistant Professor of Radiology, Harvard Medical School, Department
of Radiology, Massachusetts General Hospital, Boston, MA, USA
Contributors xix

Allison Gillams, MD
Senior Lecturer, University College of London Medical School, Honorary
Consultant, Imaging, Middlesex Hospital, London, UK

Matthew P. Goetz, MD
Assistant Professor of Oncology, Mayo Clinic College of Medicine,
Rochester, MN, USA

S. Nahum Goldberg, MD
Associate Professor of Radiology, Harvard Medical School, Director,
Abdominal Intervention and Tumor Ablation, Director, Minimally Inva-
sive Tumor Therapy Laboratory, Department of Radiology and Abdom-
inal Imaging, Beth Israel Deaconess Medical Center, Boston, MA, USA

Alan M. Harvey, MD, MBA


Director of Quality Assurance/Quality Improvement, Department
of Anesthesiology, Perioperative and Pain Medicine, Brigham and
Women’s Hospital, Boston, MA, USA

John V. Heymach, MD, PhD


Assistant Professor of Medicine, Harvard Medical School, Children’s
Hospital, Boston, MA, USA

Kullervo Hynynen, PhD


Professor of Radiology, Harvard Medical School, Director of Therapeu-
tic Research, Department of Radiology, Brigham and Women’s Hospi-
tal, Boston, MA, USA

David A. Iannitti, MD
Assistant Professor of Surgery, Brown University, Department of
Surgery, Rhode Island Hospital, Providence, RI, USA

Tiziana Ierace, MD
Department of Radiology, Ospedale Civile Busto Arsizio, Busto Arsizio,
Italy

David A. Israel, MD, PhD


Instructor in Radiology, Harvard Medical School, Staff Radiologist,
Dana-Farber Cancer Institute, Boston, MA, USA

Sapna K. Jain, MD
Resident in Radiology, University of California, San Francisco, Depart-
ment of Radiology, University Hospital, San Francisco, CA, USA

Pasi A. Jänne, MD
Assistant Professor of Medicine, Harvard Medical School, Department
of Medical Oncology, Dana-Farber Cancer Center, Boston, MA, USA
xx Contributors

Bruce E. Johnson, MD
Professor of Medicine, Harvard Medical School, Director of Thoracic
Oncology, Department of Medical Oncology, Dana-Farber Cancer
Center, Boston, MA, USA

Ferenc A. Jolesz, MD
B. Leonard Holman Professor of Radiology, Harvard Medical School,
Vice Chairman for Research, Director, Division of MRI and Image
Guided Therapy Program, Department of Radiology, Brigham and
Women’s Hospital, Boston, MA, USA

Anthony Kam, MD
Staff Clinician, Diagnostic Radiology Department, Clinical Center,
National Institutes of Health, Bethesda, MD, USA

Robert A. Kane, MD, FACR


Professor of Radiology, Harvard Medical School, Associate Chief of
Administration, Director of Ultrasound, Department of Radiology, Beth
Israel Deaconess Medical Center, Boston, MA, USA

Lily Y. Kernagis, MD
Instructor in Radiology, University of Pennsylvania, Department of
Radiology, University Hospital, Philadelphia, PA, USA

Matthew Kulke, MD
Assistant Professor of Medical Oncology, Dana-Farber Cancer Institute,
Harvard Medical School, Boston, MA, USA

Stanislav Lechpammer, MD, PhD


Department of Orthopedic Surgery, Brigham and Women’s Hospital,
Boston, MA, USA

Fred T. Lee, Jr., MD


Professor of Radiology, University of Wisconsin, Chief, Division of
Abdominal Imaging, Department of Radiology, University Hospital,
Madison, WI, USA

William R. Lees, MD
Professor of Medical Imaging, University College of London Medical
School, Department of Medical Imaging, Middlesex Hospital, London,
UK

Riccardo Lencioni, MD
Associate Professor of Radiology, University of Pisa, Department of
Oncology, Transplants, and Advanced Technologies in Medicine, Uni-
versity of Pisa, Pisa, Italy

Jonathan S. Lewin, MD
Martin W. Donner Professor and Chairman, The Johns Hopkins School
of Medicine, Radiologist-in-Chief, The Russell H. Morgan Department
Contributors xxi

of Radiology and Radiological Science, The Johns Hopkins Hospital,


Baltimore, MD, USA

Tito Livraghi, MD
Chairman Consultant, Department of Interventional Radiology,
Ospedale Civile Vimercate, Vimercate-Milano, Italy

David M. Lloyd, MBBS, FRCS (Lond.), MD


Honorary Lecturer in Medicine, Consultant Hepatobiliary and Laparo-
scopic Surgeon, Department of Surgery, Leicester Royal Infirmary,
Leicester, UK

Nathan MacDonold, MD
Department of Radiology, Brigham and Women’s Hospital, Boston, MA,
USA

Martin Mack, MD, PhD


Deputy Medical Director, Institute for Diagnostic and Interventional
Radiology, Department of Radiology, University Hospital, Frankfurt,
Germany

John P. McGahan, MD
Professor of Radiology, University of California, Davis, Director of
Ultrasound, University of California, Davis Medical Center, Sacramento,
CA, USA

Maria Franca Meloni, MD


Radiologist, Department of Interventional Radiology, Ospedale Civile
Vimercate, Vimercate-Milano, Italy

Elmar M. Merkle, MD
Associate Professor, Duke University School of Medicine, Director of
Body MRI, Department of Radiology, Duke University Hospital and
Medical Center, Durham, NC, USA

Paul R. Morrison, MS
Medical Physicist, Harvard Medical School, Department of Radiology,
Brigham and Women’s Hospital, Boston, MA, USA

Koenraad J. Mortele, MD
Assistant Professor of Radiology, Harvard Medical School, Staff
Radiologist, Department of Radiology, Brigham and Women’s Hospital,
Boston, MA, USA

Peter R. Müeller, MD
Professor of Radiology, Harvard Medical School, Director of Abdomi-
nal Imaging and Interventional Radiology, Department of Radiology,
Massachusetts General Hospital, Boston, MA, USA
xxii Contributors

David M. Nagorney, MD
Consultant, Division of Gastroenterologic and General Surgery, Mayo
Clinic; Professor of Surgery, Mayo Clinic College of Medicine,
Rochester, MN, USA

Ziv Neeman, MD
Staff Clinician, Diagnostic Radiology Department, Clinical Center,
National Institutes of Health, Bethesda, MD, USA

Sherif G. Nour, MD
Assistant Professor, Case Western Reserve University, Director of
Interventional MRI, Research, Department of Radiology, Case Western
Reserve University and University Hospitals of Cleveland, Cleveland,
OH, USA

Masao Omata, MD
Department of Gastroenterology, University of Tokyo, Tokyo

Gary Onik, MD
Director of Surgical Imaging, Center for Surgical Advancement, Cele-
bration Health/Florida Hospital, Celebration, FL, USA

Hugue Ouellette, MD
Department of Radiology, Massachusetts General Hospital, Boston,
MA, USA

Florencia G. Que, MD
Consultant, Division of Gastroenterologic and General Surgery, Mayo
Clinic; Assistant Professor of Surgery, Mayo Clinic College of Medicine,
Rochester, MN, USA

John C. Rewcastle, PhD


Adjuvant Assistant Professor, University of Calgary, Department of
Radiology, Chief Scientific Officer, Sanarus Medical, Pleasanton, CA,
USA

Daniel I. Rosenthal, MD
Professor of Radiology, Harvard Medical School, Vice Chairman,
Department of Radiology, Massachusetts General Hospital, Boston,
MA, USA

Joseph Rubin, MD
Professor of Oncology, Mayo Clinic College of Medicine, Rochester,
MA, USA

Toshihito Seki, MD
Professor of Medicine, Kansai Medical University, Division of Hepatol-
ogy, Third Department of Internal Medicine, Kansai Medical University,
Osaka, Japan
Contributors xxiii

Sridhar Shankar, MD
Assistant Professor of Radiology, University of Massachusetts, Worces-
ter, Department of Radiology, University of Massachusetts Medical
Center, Worcester, MA, USA

William E. Shiels II, DO


Associate Professor of Radiology, Ohio State University College of Med-
icine, Chairman of Radiology, Columbus Children’s Hospital, Columbus,
OH, USA

Shuichiro Shiina, MD
Associate Professor of Medicine, University of Tokyo, Department of
Gastroenterology, University Hospital, Tokyo, Japan

Stuart G. Silverman, MD
Associate Professor of Radiology, Harvard Medical School, Director,
Abdominal Imaging and Intervention, Director, CT Scan, Director,
Cross-sectional Interventional Service, Brigham and Women’s Hospital,
Boston, MA, USA

Luigi Solbiati, MD
Chairman, Department of Radiology, Ospedale Civile Busto Arsizio,
Busto Arsizio, Italy

Michael C. Soulen, MD
Professor of Radiology and Surgery, University of Pennsylvania,
Department of Radiology, University Hospital, Philadelphia, PA, USA

Elizabeth A. Stewart, MD
Associate Professor of Obstetrics, Gynecology, and Reproductive
Biology, Harvard Medical School, Clinical Director, The Center for
Uterine Fibroids, Brigham and Women’s Hospital, Boston, MA, USA

Ralf Straub, MD
Institute for Diagnostic and Interventional Radiology, Department of
Radiology, University Hospital, Frankfurt, Germany

Andrew D. Strickland, MD, MRCS


Specialist Registar in Surgery, Department of Surgery, Leicester Royal
Infirmary, Leicester, UK

Clare Tempany, MD
Professor of Radiology, Harvard Medical School, Director of Clinical
MRI and Clinical Focused Ultrasound, Department of Radiology,
Brigham and Women’s Hospital, Boston, MA, USA

Takuma Teratani, MD
Department of Gastroenterology, University of Tokyo, Tokyo
xxiv Contributors

Massimo Tonolini, MD
Department of Radiology, Ospedale Civile Busto Arsizio, Busto Arsizio,
Italy

Kemal Tuncali, MD
Instructor in Radiology, Harvard Medical School, Department of Radi-
ology, Brigham and Women’s Hospital, Boston, MA, USA

Annick D. Van den Abbeele, MD


Acting Clinical Director of Radiology, Director of Nuclear Medicine,
Dana-Farber Cancer Institute, Director, Nuclear Medicine Oncologic
Program, Brigham and Women’s Hospital, Boston, MA, USA

Vanessa A. van Raalte, MD


Victoria Radiology Consulting Group, Victoria, BC, Canada

Eric vanSonnenberg, MD
formerly: Chief of Radiology, Dana-Farber Cancer Institute, Harvard
Medical School, Interventional Radiologist, Brigham and Women’s
Hospital, Children’s Hospital, Boston, MA, USA

Thomas J. Vogl, MD, PhD


Professor, Johann Wolfgang Goethe University, Chairman of Radiology,
Institute for Diagnostic and Interventional Radiology, Department of
Radiology, University Hospital, Frankfurt, Germany

Sharon M. Weber, MD
Associate Professor of Surgery, University of Wisconsin, Department of
Surgery, University Hospital, Madison, WI, USA

Bradford J. Wood, MD
Senior Clinical Investigator and Director, Interventional Radiology
Research, Adjunct Investigator, Diagnostic Radiology Department,
Clinical Center, National Institutes of Health, Bethesda, MD, USA

Robert L. Worthington-Kirsch, MD, FSCVIR, FASA


Clinical Professor of Radiology, Philadelphia College of Osteopathic
Medicine, President, Image Guided Surgery Associates, PC, Director,
Section of Interventional Radiology, Tenet Roxborough Memorial Hos-
pital, Philadelphia, PA, USA

Stefan Zangos, MD
Institute for Diagnostic and Interventional Radiology, Department of
Radiology, University Hospital, Frankfurt, Germany
Section I
Introduction to Ablation
1
History of Ablation
John P. McGahan and Vanessa A. van Raalte

Numerous techniques have been developed for RF current. A pulsed or damped current would
tissue ablation. Techniques to kill tumor cells cauterize tissue, whereas a more continuous
include heating, freezing, radiation, chemother- current could be used to cut through tissue. The
apy, occluding the tumor blood supply, injection amount of tissue cauterized was limited to
of caustic agents directly into the tumor, as well only a few millimeters because charred tissue
as various combinations of these. While most of adhered to the tip of the knife. This first-
these were introduced in the late 20th century, generation Bovie knife was a monopolar elec-
at least one dates back to the 19th century. This trode similar to that used for contemporary
chapter reviews the general historical perspec- percutaneous RF techniques. Consequently, the
tives of these different methods, with particular grounding pads that were used were placed on
emphasis on radiofrequency ablation. the patient in a fashion similar to that employed
with most modern percutaneous RF tech-
niques. The current passing through the Bovie
Radiofrequency Ablation knife into the body is ultimately dispersed over
the wide area provided by the grounding pads.
While percutaneous methods of radiofre- The fact that RF works by causing ionic agi-
quency ablation (RFA) are relatively new, the tation of the tissues surrounding the needle was
basic technique for RFA was described over a first demonstrated by Organ (5). The shaft of
century ago by D’Arsonval (1), who, in 1891, the needle does not produce heat. Rather, the
first demonstrated that when RF waves passed heat is produced in the tissues, and that leads to
through tissue, they caused an increase in tissue coagulation and cellular necrosis. Fairly rapid
temperature. In the early 1900s, RF was used application of current leads to a small region of
in relatively few medical applications (2,3). In coagulation as well as local tissue charring.
1910 Beer (2) described a new method for the Charring acts to inhibit further ionic agitation,
treatment of bladder neoplasms using cauteri- thus limiting the amount of surrounding coag-
zation through a cystoscope. In 1911 Clark (3) ulation necrosis. In 1990, two independent
described the use of oscillatory dessication in investigators used a modification of prior RF
the treatment of malignant tumors that were techniques to create coagulation necrosis that
accessible for minor surgical procedures. could be applied via the percutaneous route.
However, RF for medical applications was McGahan et al (6) described their research in
not widely popularized until after the introduc- the English literature, and in the same year
tion of the Bovie knife in 1928 by Cushing and Rossi et al (7) described a similar technique
Bovie (4) (Liebel Florsheim, Cincinnati, Ohio). in the Italian literature. These investigators
This instrument could be used either for cau- replaced the Bovie knife with specially de-
terization or for cutting tissue by varying the signed needles insulated to the distal tip. This

3
4 J.P. McGahan and V.A. van Raalte

needle placement and to assess the echogenic


response in the tissue surrounding the RF
needle during ablation. They documented
increased echogencity surrounding the exposed
needle tip following the application of current
(Fig. 1.2). This echogenic response was ellipsoid
in appearance and roughly corresponded to the
volume of coagulation necrosis seen on patho-
logic examination. There was a central zone of
char around the needle tip and an accompany-
ing larger zone of coagulation necrosis. There
A
was also a zone of hemorrhage at the interface
between the coagulated tissue and normal liver
(Fig. 1.3).
In 1993 this technique was used for RFA of
liver tumors in humans (9). It was soon investi-
gated by others and shortly thereafter became
commercially available. RF generators and
needles were developed that could be used for
percutaneous, laparoscopic, or open ablation. A
problem with initial needle design is that the
volume of tissue necrosis was limited to approx-
imately 2 cm3 when using an uninsulated needle
length of approximately 2 cm. Multiple applica-
B
tions with this type of needle could ablate a
Figure 1.1. (A) Original needle design. Photograph 2-cm-diameter tumor, with a volume slightly
of original needle design shows standard stock greater than 4 cm3 (Fig. 1.4). The lesion was
needle that is insulated (arrow) up to distal tip. elongate, with the long axis of the necrosis par-
Needle tip is not insulated. (B) Original needle allel to the shaft of the needle. However, most
design. Drawing shows theoretic lesion that would be lesions that are treatable by RF are oval rather
produced if noninsulated needle tip were used than elongate in shape, and smaller lesions are
during monopolar radiofrequency electrocautery.
the exception rather than the rule. To ablate a
(Reprinted with permission from The American
lesion of approximately 4 cm in diameter with
Journal of Roentgenology.)
a tumor volume greater than 36 cm3, one would
need at least 18 separate optimal placements of
the originally designed needle to eradicate the
design directed the flow of current into the tumor completely. Even more treatments
target tissue. Thus, an insulated needle could be would be needed if a volume of normal tissue
percutaneously placed deep into a target organ had to be destroyed to ensure tumor-free
such as the liver to produce an area of focal margins and prevent recurrence.
necrosis (Fig. 1.1). The length of the central axis There have been a number of commercially
of the lesion could be increased by exposing the available technical advances intended to solve
tissue to a larger area of uninsulated needle. A the problem of the limited volume of tissue
limitation of this original needle design was necrosis associated with percutaneous RF
that the diameter of the lesion to be ablated needles. Early RF generators for percutaneous
needed to be 1.5 cm or less. Grounding pads applications produced a maximum of 50 W of
were placed in a similar manner to that used for power, whereas modern generators can
the Bovie knife. produce between 150 and 200 W. Recent
In 1992 McGahan et al (8) showed that ultra- improvements in software designs and methods
sound could be used both to monitor the RF of power deposition ensure maximal energy
A

Figure 1.3. In vivo histologic correlation for radio-


frequency coagulation of swine liver. Photograph of
in vivo liver reveals central area of charred tissue (1)
surrounded by coagulative necrosis (2) and hyper-
emic rim (arrow, 3). L, healthy liver. (Reprinted with
permission from J.P. McGahan, J.M. Brock, H. Tesluk
et al. “Hepatic ablation with use of radiofrequency
electocautery in the animal model.” JVIR 19923:
291–297.)
B

C
Figure 1.2. In vitro radiofrequency coagulation of
bovine liver. (A) Needle being placed under ultra-
sound guidance showing uninsulated needle tip
(arrowhead 5). (B,C) With increasing power deposi- Figure 1.4. Overlapping coagulation. One method
tion, there is increased echogenicity of liver sur- of increasing coagulation is to produce multiple
rounding the needle tip (curved arrow). regions of necrosis by repositioning the needle.
5
6 J.P. McGahan and V.A. van Raalte

the generators, the regions of coagulation sur-


rounding each prong coalesce, resulting in
increased volumes of necrosis. Other needle
designs enable the operator to deploy an even
greater number of prongs from the needle tip,
producing a larger uniform volume of necrosis
(Fig. 1.6).
In 1996 and 1997 Goldberg et al (11) and
Lorentzen et al (12) described an innovative
approach in which the needle tip was cooled by
chilled saline pumped through the shaft of the
needle. While it may seem counterintuitive
to cool the needle tip in an attempt to create
Figure 1.5. Some of the original self-expanding a larger region of coagulation necrosis from
needles had only a few prongs that deployed. This tissue heating, these investigators showed that
could lead to an uneven distribution of coagulation cooling of the needle tip prevented tissue
necrosis.
charring that occurred with the original needle
designs by McGahan and Rossi. A cooled
transmission to the target organ before discon- needle allows for a decrease in the temperature
tinuance of current flow and coagulation. of the tissue surrounding the needle tip, thereby
Generally, the greater the amount of power producing tissue coagulation without signifi-
disposition to the target area, the greater the cant charring. The reduction in charring results
volume of tissue destroyed. Additionally, in further ionic agitation with increased volume
several different types of needles have been of tissue necrosis. A variation of this technology
designed to increase the volume of tissue necro- utilizes three or more cooled tip needles
sis. In 1997 LeVeen (10) described an approach (“clustered”) to increase the volume of tissue
for a monopolar needle or electrode that had necrosis (13) (Fig. 1.7).
separate prongs deployable from the needle tip. Further research has shown that several
Each of these prongs is uninsulated and func- factors can alter the amount of tissue necrosis.
tions as an RF antenna for dispersion of current Coagulation necrosis is a function of the total
(Fig. 1.5). By increasing the energy output of RF energy deposited into the tissue, multiplied

A B
Figure 1.6. Photographs of radiofrequency needle prongs protrude in umbrella configuration from tip
designs. (A) Prongs protrude in “Christmas tree” of needle manufactured by Radiotherapeutics.
configuration from tip of needle manufactured by (Boston Scientific, Natick, MA.)
RITA Medical Systems, Mountain View, CA. (B) Ten
1. History of Ablation 7

For instance, rapid application of current


creates charring that reduces the water content
of the tissue and ionic agitation, thereby limit-
ing the amount of tissue necrosis.
In 1997 Livraghi et al (17) performed RF
application on animal liver both ex vivo and in
vivo. Their studies were conducted both with
and without intraparenchymal saline injection.
Radiofrequency application with continuous
saline infusion also was evaluated. Livraghi et al
found the largest volume of coagulation necro-
sis resulted from RF performed with continuous
saline perfusion, while the smallest volumes
were associated with RF performed without
saline. Based on these data, they performed con-
tinuous saline-enhanced RF in 14 patients with
liver metastases and one patient with primary
cholangiocarcinoma. They documented com-
plete necrosis in 13 of 25 liver lesions with
diameters up to 3.9 cm.They concluded that this
continuous saline infusion technique had
Figure 1.7. Photographs of radiofrequency needle promise in RF treatment of larger tumors.
designs. Single (A) and clustered (B), cooled-tip Curley and Hamilton (18) increased coagula-
needles manufactured by Valleylab. (Photos printed tion diameters from 1.4 to 2.6 cm by infusion of
with permission from Valleylab.) 10 mL/min of normal saline into ex vivo liver for
4 minutes during RF application. Miao et al (19)
by local tissue interactions, minus the local and achieved coagulation diameters up to 5.5 cm by
systemic heat lost (14–16). Modern RF gen- infusion of 1 mL/min of hypertonic saline into ex
erators with higher wattages can increase the vivo liver for 12 minutes. As a direct conse-
power deposition with a resulting increase in quence of this research and that performed by
tissue necrosis. Local tissue characteristics and other investigators, saline-perfused RF has
tissue interaction with RF current are impor- become a promising treatment option in man-
tant parameters in creating regions in necrosis. agement of liver tumors (20) (Fig. 1.8).

A B
Figure 1.8. (A) Diagram of needle design HITT RF needle with channels for dispersion of saline during
ablation needle in which saline is infused through RF application (BerCHTOLD Medical Electronic,
infusion port at proximal end. (B) Distal end of Tuttlingen, Germany).
8 J.P. McGahan and V.A. van Raalte

Optimal ablation results are achieved when Clinical results using the Pringle maneuver
local temperature is maintained between 60° are reported to be excellent. In 1999 Curley
and 100°C, with nearly instantaneous tissue et al (24) reported the results of RF in 123
coagulation necrosis (14–16). Lower tempera- patients. The Pringle maneuver was used in the
tures require longer exposure time to achieve majority of the 92 patients who were treated
the desired result. For example, at 46°C, at least with RF intraoperatively. Tumor recurrence
1 hour is required to ensure cell damage. Con- was detected in only three of the 169 lesions
versely, a rapid increase in local tissue temper- that were treated at a median follow-up of 15
ature above 100°C causes tissue charring that months (24).
limits effective coagulation necrosis. Some
manufacturers have devices in the needle tips
Chemotherapy and RF
that measure local tissue temperature during
RF application. There has been considerable interest in
combining RF with chemotherapy. In 1999
Kainuma et al (25) combined intraarterial infu-
Decreased Blood Flow and RF sion of chemotherapy with RF electrocautery
To compensate for the “heat sink” effect of to treat hepatic metastases from colon cancer.
large vessels within tumors, several investi- Other investigators have shown there is an
gators have described methods intended to enhanced effect when chemotherapeutic agents
decrease blood flow to the organ of interest, are heated to hyperthermic temperatures
before or during RF application. This approach between 42° and 45°C prior to infusion into
was first presented by Buscarini et al (21), the target tissue (26). In an initial report by
who, in 1999, described the use of RF combined Goldberg et al (27), increased coagulation
with transcatheter arterial embolization for necrosis was documented when RFA was com-
treatment of large hepatocellular carcinomas bined with intratumoral injection of doxyru-
(HCC). Rossi et al (22) used balloon occlusion bicin. The volume of necrosis with the injection
of the hepatic artery during RFA to treat larger of doxyrubicin alone was 2 to 3 mm in an R3230
HCCs. In 81% of HCCs averaging 4.7 cm in mammary adenocarcinoma model, whereas
size treated by Rossi et al, there was no local RFA alone produced 6.7 mm of necrosis. When
recurrence at 1 year after this combined RF was combined with injection of doxorubicin,
therapy. a necrosis diameter of 11.4 mm was achieved
Another method of decreasing the blood (27), a marked improvement in monotherapy.
supply to hepatic tumors is use of the Pringle
maneuver, which is performed during open
surgical application of RF. After the RF needle
Summary
is positioned within the tumor, the surgeon Over the past 10 years there has been a rapid
temporarily occludes the portal vein and the advancement in the utilization of RF electro-
hepatic artery, thereby temporarily interrupting cautery for percutaneous, laparoscopic, and
the blood supply to the liver, which significantly open surgical applications in the treatment of a
reduces the “heat sink” effect. Goldberg et al number of different tumors. It was only in the
(23) used this technique in a limited series early 1990s that McGahan et al and Rossi et al
reported in 1998. They were able to achieve an independently described the pioneering appli-
increase in the volume of the tumor necrosis in cation of the percutaneous use of RF energy in
patients when employing the Pringle maneuver the animal liver model. Over the past 10 years,
compared to patients being treated with similar manufacturers have designed more powerful
RF parameters but without the Pringle maneu- generators, developed special programs for
ver. The tumor necrosis diameter without the heat deposition, and achieved improved needle
Pringle maneuver was 2.5 cm compared with designs that enable the creation of larger
4.1 cm when using this maneuver. volumes of tissue necrosis. Most recently, a
1. History of Ablation 9

number of investigators have explored other were treated with percutaneous injection of
methods for increasing the amount of RF- 95% ethanol into tumors that were less than
induced tissue necrosis including catheter arte- 4 cm in diameter. From three to nine sessions
rial occlusion during performance of RF, as well were performed for each lesion, according
as intraoperative performance of the Pringle to size. This work established the feasibility
maneuver. Others have used chemotherapy in and value of percutaneous intratumoral alco-
combination with RF to increase the volume hol injection to treat selected small hepatic
of tumor ablated. Some investigators have neoplasms.
demonstrated that injection of saline into the In 1988, Livraghi et al (32) published findings
tumor during RF application can increase involving a series of 23 patients with HCC with
tumor necrosis. There is little doubt that RF tumor diameters less than 4.5 cm; 32 lesions
alone or in combination with other technolo- were treated by ethanol injections performed
gies will find further utilization and wider appli- under percutaneous ultrasound guidance. A
cation in percutaneous treatment of tumors and total of 271 treatment sessions achieved a 1-
other diseases throughout the body. year survival rate in 12 patients of 92%. All the
lesions were smaller at 6 to 27 months' follow-
up. In the early 1990s, other authors began using
Percutaneous Ethanol Injection intratumoral ethanol for ablation of liver
tumors. These efforts established the efficacy of
Percutaneous tissue ablation using ethanol is percutaneous ethanol injection (PEl) in the
not new. One of the first descriptions of its use treatment of hepatocellular carcinoma (33,34).
was in the treatment of renal cysts by Bean (28) In 1991, Livraghi et al (35) demonstrated that
in 1981. He treated 34 benign renal cysts in 29 while PEl could be used to treat focal metasta-
patients using direct injection of 95% ethanol. tic disease, its benefits are limited by the natural
The technique used by Bean more than 20 years course of the disease.
ago has not changed significantly to the present. In 1995 Livraghi et al (36) demonstrated PEl
After confirming with contrast fluoroscopy that to be safe, effective,.low cost, and easily repro-
there was no leakage from a benign cyst, 4.6- ducible in the treatment of patients with HCC.
French polyethylene catheters were pla'ced into They performed this technique in 746 patients
the cyst. Approximately 3% to 44% of the cyst with HCC. The survival rates of patients were
volumes were replaced with 95% ethanol that generally good, but dependent on the severity
was maintained in the cyst for 10 to 20 minutes. of accompanying cirrhosis. Patients with Child's
In this series, there was only one recurrence at A cirrhosis had a much better survival than
3 months. During the past two decades, other their counterparts with Child's B or C cirrhosis.
agents have been used to sclerose renal cysts. In Livraghi et al believed that the survival after
1985 Bean and Rodan (29) described a similar PEl was comparable to that of surgery, in part
technique for treatment of hepatic cysts. due to the increased mortality from surgery and
In 1985 Solbiati et al (30) injected absolute liver damage associated with resection.
alcohol into enlarged parathyroids in patients
with secondary hyperparathyroidism. This tech-
nique was useful in reducing the size of the Other Injectables
parathyroid glands in those patients in whom
surgery was contraindicated. The following Other injected liquids have been used to treat
year, Livraghi et al (31) described the use of tumors. In 1986 Livraghi et al (37) performed
ultrasound-guided needle placement into 14 what they called percutaneous intratumoral
hepatic lesions. These included nine patients chemotherapy (PIC) under ultrasound guid-
with HCC, four with hepatic metastasis from ance in 12 selected patients with tumors that
gastric carcinoma, and one peritoneal metasta- had been unresponsive to conventional treat-
sis from transitional cell carcinoma, All patients ment. They injected a single agent or a combi-
10 IP. McGahan and v.A. van Raalte

nation of either 5-fluorouracil, methotrexate, or bleeding and minimal damage to adjacent


cyclophosphamide depending on the histology tissue (44). In 1986 in the Japanese literature,
of the tumor. There were 119 sessions of PIC in Fujishima et al (45) published an experiment
which the authors achieved either partial or using the Nd:YAG laser as a possible treatment
total pain control and stable disease response of deep-seated brain tumors. Hashimoto (46)
in 60% of the patients. During that same year, published an experiment on the clinical appli-
Livraghi et al (38) described the treatment cation of the effects of lasers for hyperthermia
of inoperable HCCs smaller than 3cm with of liver neoplasms. In 1998 Tajiri et al (47)
percutaneous interstitial chemotherapy using induced local interstitial hyperthermia using
5-fluorouracil injected under ultrasound guid- the Nd:YAG laser to treat gastrointestinal and
ance. They documented some fibrosis in the pancreatic carcinomas that had been trans-
region of treatment at 3 months, and they sug- planted into mice. These authors demonstrated
gested that this type of therapy could be useful there were areas of ischemic infarction and
as an alternative treatment for HCC. marked necrosis of the pancreatic tumor in the
In the early 1990s Honda and his coauthors region of laser treatment.
advocated use of percutaneous hot saline injec- In 1990 Hahl et al (48) treated seven patients
tion therapy (PSITN) as an alternative to with malignant tumors using the Nd:YAG laser
percutaneous ethanol treatment for hepatic during open laparotomy. They used sapphire
tumors. One hundred twenty-three patients probes with the power settings between 6 and
with HCC were treated with PSITN. A thera- 8W. Even though only limited necrosis was
peutic effect occurred in all patients. Even produced by these lasers, they believed that
so, hot saline therapy did not become widely selective destruction of malignant tumors was
utilized. possible using laser-induced hyperthermia. In
Another percutaneous therapy advocated in 1991 Van Hillegersberg et al (49) studied the
the 1990s was injection of acetic acid under use of laser coagulation in rat liver metastases
sonographic guidance (41-43). For example, in and again demonstrated selective necrosis
1994 Ohnishi et al (41) performed ultrasound- using this technique. In 1992 Dowlatshahi et al
guided percutaneous acetic acid injection (PAl) (50) reported in situ focal hyperthermia that
in the treatment of hepatocellular carcinoma. was generated by an Nd:YAG laser. The laser
PAl with 50% acetic acid was performed in 25 was introduced through a 19-9auge needle
patients with solitary HCCs 3 cm or less in inserted percutaneously into the liver under
diameter. There was a total of 82 sessions with ultrasound guidance. This process was moni-
no significant complications and no evidence of tored under ultrasound and confirmed by com-
viable HCC on follow-up biopsy. The survival puted tomography. Tumor growth was halted
rate among their 23 patients was 100% at 1 year at 3 months, indicating partial response in
and 92% at 2 years. In the future, research into humans.
these and other injectable agents for the treat- In 1993 Nolsoe et al (51) used laser hyper-
ment of focal neoplasms seems likely. thermia in 11 patients with 16 colorectal liver
metastases. Real-time ultrasound was utilized
to monitor this technique. They demonstrated
Lasers in this larger patient group that laser hyper-
thermia was feasible, effective, and safe. Fol-
A neodymium: yttrium-aluminum-garnet (Nd: lowing that publication, others have obtained
YAG) laser system with ceramic rods was ini- wider experience using these techniques in clin-
tially developed and tested in animals, then ical practice in the treatment of primary and
later used in clinical practice to treat head and secondary liver tumors. Some investigators
neck tumors. This method was initially used to have used interstitial laser hyperthermia to
produce an "incision," rather than for tumor treat neck tumors, in various neurosurgical
destruction. It was thought to be a precise applications, and in limited animal experimen-
method of surgical dissection, with reduced tation with mammary tumors (52-54).
1. History of Ablation 11

Microwave Hyperthermia on the volume of tissue necrosis that can be


achieved utilizing microwave. Improvements in
Compared to other techniques, percutaneous needle design and technology should address
microwave coagulation of tumors arrived some- this problem.
what late on the scene. In 1994 Seki et al (55)
described a single case of percutaneous trans-
hepatic microwave coagulation therapy for a Cryotherapy
patient with HCC with bile duct involvement.
That same year, they described how percuta- Some of the initial work in cryotherapy was
neous microwave coagulation therapy (PMCT) undertaken in the late 1970s, and most of it was
was used in the local treatment of unresectable experimental (62-66). In 1987 Ravikumar and
HCC (56). This was performed in 18 patients his associates (67) reported their surgical expe-
with small diameter (less than 2cm) HCCs. A rience with hepatic cryosurgery for metastatic
microwave electrode 1.6mm in thickness was colon cancer to the liver. Their development of
inserted percutaneously under ultrasound 8 to 12mL liquid nitrogen-cooled probes, suit-
guidance into the tumor region. The microwave able for surgical placement within the liver,
was generated using 60W for 120 seconds. As established the advent of interstitial hepatic
with other thermal techniques, an echogenic cryosurgery. Real-time ultrasound was used to
response around the tumor was detected on verify the extent of treatment and to measure
ultrasound. Complete necrosis of the tumor the size of the increasing iceball that was
area was documented in one patient who under- created by freezing. Freezing and thawing for
went hepatectomy after PMCT. In a short three cycles was an effective means of ensur-
follow-up period lasting from 11 to 33 months, ing tumor necrosis. The average size of these
the patients had no local recurrence following lesions was in the range of 3cm. Further exper-
treatment of these tumors. imentation by Onik et al (68) in 1995 confirmed
In 1995 Murakami et al (57) used PMCT in the animal model the efficacy of sonography
therapy in nine HCCs less than 3cm in diame- for monitoring hepatic cryotherapy.
ter, and it was found to be a useful alternative By the late 1980s and early 1990s, investiga-
to other forms of interstitial therapy for treat- tors such as Charnley et al (69) and Onik et al
ment of small HCCs. Hamazoe et al (58) also (68) demonstrated the usefulness of this tech-
reported the use of microwave tissue coagula- nique. However, the disadvantages of cryother-
tion (MTC) during laparotomy in eight patients apy included that the technique required open
with nonresectable HCCs. They found that surgery and that fairly large probes had to be
intraoperative MTC appeared to be an effec- placed into the lesions. Consequently, the sur-
tive method for inducing tumor necrosis that gical risks must be factored into analysis of
could be used in combination with local surgi- morbidity and mortality for this patient group.
cal resection for multiple HCCs when radical In addition to the usual pleural effusions and
liver resection was not feasible. the morbidity associated with the open tech-
In 1996 others reported using microwave nique, there were certain unique complications
hyperthermia guided by ultrasound in the per- or imaging findings that occurred specifically
cutaneous treatment of HCCs. These included with cryosurgery. These include posttreatment
limited results in 12 patients reported by Dong "cracking" of the hepatic parenchyma, air or
et al (59), two patients with long-term survival hemorrhage in the lesion, and subcapsular hem-
reported by Sato et al (60), and eight patients orrhage. Smaller percutaneous cryotherapy
treated by Yamanaka et al (61). The patients probes have been developed that eliminate the
in the Yamanaka series were treated both risk associated with open surgical exploration
under laparoscopic and open methods. More (68). The techniques and various applications
recent and larger series describe the utility of of modern cryotherapy will be explained in
microwave hyperthermia in the treatment of further detail in the accompanying chapter on
focal masses. Currently there are limitations that topic.
12 J.P. McGahan and V.A. van Raalte

High-Intensity Focused This technique for the ablation ofliver tumors


is not without its challenges. For example,
Ultrasound the movement of the liver associated with
Documenting the first published research on respiration will require gating of HIFU for
the cavitation effects of ultrasound has been transcutaneous applications to ensure that the
difficult. High-intensity focused ultrasound wastarget lesion remains within the zone of
described in 1989 as a method to treat discrete maximum intensity of the ultrasound beam to
liver tumors. This was investigated in both in minimize collateral damage to nonneoplastic
vivo and excised liver samples by ter Harr and tissue.
associates (72). They performed both qualita- Where organ movement is not a problem,
tive and quantitative studies to demonstrate HIFU has been more successful. For example,
when used in the prostate or when used intra-
the feasibility of this technique, with particular
attention devoted to the lesion shape, position,operatively, the ultrasound can be directly
and volume in relation to the duration of the applied to the organ of interest and kept in one
ultrasound exposure. position for the duration of HIFU application.
There has been research involving HIFU appli-
Further research in this area was carried out
in the early 1990s by Chapelon et al (73), Prat cations in the genitourinary system including
et al (74), and Yang et al (75). At least two ofablation of renal and prostatic lesions (79-81).
these investigators reviewed the use of high- Gelet et al (80) demonstrated the utility of
intensity focused ultrasound (HIFU) in the HIFU to induce coagulation necrosis in the
treatment of hepatic lesions. One study (75) canine prostate. They determined the different
examined the use of HIFU to treat hepatomas levels of HIFU required to achieve varying
amounts of tissue ablation within the prostate.
implanted into the livers of rats. A total of 112
rats with liver tumors were divided into two There was homogeneous coagulation necrosis
groups-those that had HIFU and those that within the lesions that later progressed to
did not. A significant inhibition of tumor inflammatory fibrosis and finally to sclerosis
growth of 65% was seen in the HIFU-treated with cavity formation. This study in animals
group on the third day posttreatment, with 93% demonstrated the potential for creating irre-
growth inhibition on the 28th day following versible prostate lesions using HIFU without
treatment. There was evidence of both tumor damaging the rectal wall. In the same year,
necrosis and fibrosis. This is one of the first Gelet et al (81) successfully used a similar tech-
studies on the use of HIFU for treatment of nique in nine patients with prostate hyper-
liver tumors. trophy. Finally, as was mentioned previously,
In 1992 and in 1993 Yang et al (76,77) pro- some of the problems resulting from organ
duced further work demonstrating that HIFU motion can be eliminated when HIFU is used
could destroy targeted deep-seated tissue intraoperatively. .
within the liver without causing damage to
nontargeted tissue. These experiments demon-
strated the potential usefulness of this therapy Summary
for ablation of liver cancer without the need for
laparotomy. Modern medicine is constantly evolving less
Similar experimentation was performed invasive methods for the treatment of disease.
by Sibille et al (78) in 1993. They demon- While some of this research with tissue ablation
strated that in the in-vivo rabbit liver a rela- was documented over 100 years ago, the major-
tionship exists among HIFU intensity levels, ity of investigative efforts have taken place
exposure times, and the posttreatment lesion within the past 20 years. In the future there
appearance. This work demonstrated the need will undoubtedly be additional research into
to adapt intensity to the depth of the target these techniques and the development of new
tissue. methods of tissue ablation.
1. History of Ablation 13

References 15. McGahan JP, Dodd GD III. Radiofrequency


ablation of the liver: current status. AJR 2001;
1. D'Arsonval MA. Action physiologique des 176:3-16.
courants alternatifs. C R Soc BioI 1891; 16. Zevas NT, Kuwayama A. Pathological charac-
43:283-286. teristics of experimental thermal lesions: com-
2. Beer E. Removal of neoplasms of the urinary parison of induction heating and radiofrequency
bladder: a new method employing high fre- electrocoagulation. J Neurosurg 1972;37:418-
quency (oudin) currents through a cauterizing 422.
cystoscope. JAMA 1910;54:1768-1769. 17. Livraghi T, Goldberg SN, Lazzaroni S, et al.
3. Clark WL. Oscillatory desiccation in the treat- Saline-enhanced radiofrequency tissue ablation
ment of accessible malignant growths and minor in the treatment of liver metastases. Radiology
surgical conditions. J Adv Ther 1911;29:169-183. 1997;202:205-210.
4. Cushing H, Bovie WT. Electro-surgery as an 18. Curley MG, Hamilton PS. Creation of large
aid to the removal of intracranial tumors. Surg thermal lesions in liver using saline-enhanced
Gynecol Obstet 1928;47:751-784. RF ablation. Proceedings of the 19th Interna-
5. Organ LW. Electrophysiologic principles of tional Conference IEEEIEMBS, Chicago,
radiofrequency lesion making. Appl Neuro- October 3D-November 2,1997.
physioI1976-1977;39:69-76. 19. Miao Y, Ni Y, Mulier S, et al. Ex vivo experiment
6. McGahan JP, Browing PD, Brock JM, Tesluk H. on radiofrequency liver ablation with saline infu-
Hepatic ablation using radiofrequency electro- sion through a screw-tip cannulated electrode.
cautery. Invest Radiol 1990;25:267-270. J Surg Res 1997;71:19-24.
7. Rossi S, Fornari F, Pathies C, Buscarini L. 20. Hansler J, et al. Percutaneous ultrasound guide
Thermal lesions induced by 480 KHz localized radiofrequency tissue ablation (RFTA) with per-
current field in guinea pig and pig liver. Thmori fused needle applicators-treatment of hepato-
1990;76:54-57. cellular carcinoma and liver metastases-first
8. McGahan JP, Brock JM, Tesluk H, et al. Hepatic results. Proceedings of EMBEC 1999;pt 11:1576-
ablation with use of radiofrequency electro- 1577.
cautery in the animal model. J Vasc Intervent 21. Buscarini L, Buscarini E, Stasi M, Quaretti P,
RadioI1992;3:291-297. Zangrandi A. Percutaneous radiofrequency
9. McGahan JP, Scheider P, Brock JM, Teslik H. thermal ablation combined with transcatheter
Treatment of liver tumors by percutaneous radio- arterial embolization in the treatment of large
frequency electrocautery. Semin Intervent hepatocellular carcinoma. Ultraschall Med 1999;
RadioI1993;10(2):143-149. 20:47-53.
10. LeVeen RF. Laser hyperthermia and radiofre- 22. Rossi S, Garbagnati F, Lencioni R. Unresectable
quency ablation of hepatic lesions. Semin Inter- hepatocellular carcinoma: percutaneous radio-
vent RadioI1997;14:313-324. frequency thermal ablation after occlusion of
11. Goldberg SN, Gazelle GS, Solbiati L, Rittman tumor blood supply. Radiology 2000;217:119-
WJ, Mueller PRo Radiofrequency tissue ablation: 126.
increased lesion diameter with a perfusion elec- 23. Goldberg SN, Hahn PF, Tanabe KK, et al. Per-
trode. Acad Radiol 1996;3:636-644. cutaneous radiofrequency tissue ablation: Does
12. Lorentzen T, Christensen NE, Nolsoe CP, Torp- perfusion-mediated tissue cooling limit coagula-
Pedersen ST. Radiofrequency tissue ablation tion necrosis? J Vasc Intervent Radiol 1998;9:
with a cooled needle in vitro: ultrasonography, 101-111.
dose response, and lesion temperature. Acad 24. Curley SA, Izzo F, Delrio P, et al. Radio-
Radiol 1997;4:292-297. frequency ablation of unresectable primary and
13. Goldberg SN, Solbiati L, Hahn PF, et al. Large metastatic hepatic malignancies: results in 123
volume tissue ablation with radiofrequency patients. Ann Surg 1999;230:1-8.
by using a clustered, internally cooled electrode 25. Kainuma 0, Asano T, Aoyama H, et al. Com-
technique: laboratory and clinical experience in bined therapy with radiofrequency thermal abla-
liver metastases. Radiology 1998;209:371-379. tion and intra-arterial infusion chemotherapy
14. Goldberg SN, Dupuy DE. Image-guided for hepatic metastases from colorectal cancer.
radiofrequency tumor ablation: challenges and Hepatogastroenterology 1999;46(26):1071-1077.
opportunities-part 1. J Vasc Intervent Radiol 26. Seegenschmiedt MH, Brady LW, Sauer R. Inter-
2001;12:1021-1032. stitial thermo-radiotherapy: a review on techni-
14 IP. McGahan and V.A. van Raalte

cal and clinical aspects. Am J Clin Oncol 1990; 40. Honda N, Guo Q, Uchida H, Ohishi H, Hiasa Y.
13:352-363. Percutaneous hot saline injection therapy for
27. Goldberg SN, Saldinger PF, Gazelle GS, et al. hepatic tumors: an alternative to percutaneous
Percutaneous tumor ablation: increased coagula- ethanol injection therapy. Radiology 1994;
tion necrosis with combined radiofrequency and 190(1):53-57.
percutaneous doxorubicin injection. Radiology 41. Ohnishi K, Ohyama N, Ito S, Fujiwara K. Small
2001. hepatocellular carcinoma: treatment with US-
28. Bean WI Renal cysts: treatment with alcohol. guided intratumoral injection of acetic acid.
Radiology 1981;138(2):329-331. Radiology 1994;193(3):747-752.
29. Bean WJ, Rodan BA. Hepatic cysts: treatment 42. Ohnishi K, Yoshioka H, Ito S, Fujiwara K. Treat-
with alcohol. AJR 1985;144(2):237-241. ment of nodular hepatocellular carcinoma larger
30. Solbiati L, Giangrande A, De Pra L, Bellotti E, than 3 em with ultrasound-guided percutaneous
Cantu P, Ravetto C. Percutaneous ethanol injec- acetic acid injection. Hepatology 1996;24(6):
tion of parathyroid tumors under US guidance: 1379-1385.
treatment for secondary hyperparathyroidism. 43. Ohnishi K, Nomura F, Ito S, Fujiwara K. Prog-
Radiology 1985;155(3):607-610. nosis of small hepatocellular carcinoma (less
31. Livraghi T, Festi D, Monti F, Salmi A, Vettori C. than 3cm) after percutaneous acetic acid injec-
US-guided percutaneous alcohol injection of tion: study of 91 cases. Hepatology 1996;23(5):
small hepatic and abdominal tumors. Radiology 994-1002.
1986;161(2):309-312. 44. Ohyama M, Katsuda K, Nobori T, et al. Treat-
32. Livraghi T, Salmi A, Bolondi L, Marin G, Arienti ment of head and neck tumors by contact
V, Monti F. Small hepatocellular carcinoma: Nd-YAG laser surgery. Auris Nasus Larynx
percutaneous alcohol injection-results in 23 1985;12(suppI2):S138-142.
patients. Radiology 1998;168(2):313-317. 45. Fujishima I, et al. Experimental study of deep
33. Ebara M, Otho M, Sugiura N, Okuda K, Kondo brain radiation with the Nd-YAG laser. A possi-
F, Kondo Y. Percutaneous ethanol injection for ble new treatment for deep-seated brain tumor.
the treatment of small hepatocellular carcinoma: Neurol Med Chir (Tokyo) 1986;26(8):621-627.
study of 95 patients. J Gastroenterol Hepatol 46. Hashimoto D. Clinical application of the thermal
1990;5:616-626. effect of lasers. 2. Application of the laser
34. Shiina S, Tagawa K, Unuma T, et al. Percutaneous thermal effect to the therapy of liver neoplasms.
ethanol injection therapy for hepatocellular car- Nippon Rinsho 1987;45(4):888-896.
cinoma: a histopathologic study. Cancer 1991;68: 47. Tajiri H, Oguro Y, Egawa S, Sugimura T. Local
1524-1530. hyperthermia using a low powered Nd:YAG
35. Livraghi T, Vettori C, Lazzaroni S. Liver metas- laser for pancreatic and gastric carcinoma.
tases: results of percutaneous ethanol injections Hokkaido Igaku Zasshi 1998;63(6):889-896.
in 14 patients. Radiology 1991;179(3):709-712. 48. Hahl J, Haapiainen R, Ovaska J, Puolakkainen P,
36. Livraghi T, Giorgio A, Marin G, et al. Hepato- Schroder T. Laser-induced hypothermia in the
cellular carcinoma and cirrhosis in 746 patients: treatment of liver tumors. Lasers Surg Med 1990;
long-term results of percutaneous ethanol injec- 10(4):319-321.
tion. Radiology 1995;197(1):101-108. 49. Van Hillegersberg R, Kort WJ, ten Kate FJ,
37. Livraghi T, Bajetta E, Matricardi L, Villa E, Terpstra OT. Water-jet-cooled Nd:YAG laser
Lovati R, Vettori C. Fine needle percutaneous coagulation: selective destruction of rat liver
intratumoral chemotherapy under ultrasound metastases. Lasers Surg Med 1991;11(5):445-454.
guidance: a feasibility study. Tumori 1986;72(1): 50. Dowlatshahi K, Bhattacharya AK, Silver B,
81-87. Matalon T, Williams JW. Percutaneous intersti-
38. Livraghi T, Ravetto C, Solbiati L, Suter F. Percu- tial laser therapy of a patient with recurrent
taneous interstitial chemotherapy of a small hepatoma in a transplanted liver. Surgery
hepatocellular carcinoma under ultrasound 1992;112(3):603-606.
guidance. Tumori 1986;72(5):525-527. 51. Nolsoe CP, Torp-Pedersen S, Burcharth F, et al.
39. Honda N, Guo Q, Uchida H, et al. Percutaneous Interstitial hyperthermia of colorectal liver
hot water injection therapy (PhoT) for hepatic metastases with a US-guided Nd-YAG laser with
tumors: a clinical study. Nippon Igaku Hoshasen a diffuser tip: a pilot clinical study. Radiology
Gakkai Zasshi 1993;53(7):781-789. 1993;187(2):333-337.
1. History of Ablation 15

52. Panjehpour M, Overholt BF, Frazier DL, 64. Grana L, Airan M, Gordon M, Johnson R. Cryo-
Klebanow ER. ND:YAG laser-induced hyper- genic technique for resection of carcinoma of the
thermia treatment of spontaneously occurring colon. Int Surg 1978;63(5):53-56.
veterinary head and neck tumors. Lasers Surg 65. Walzel C. Current status of cryogenic surgery.
Med 1991;11(4):351-355. [Article in German] Chirurg 1978;49(4):202-
53. Dowlatshahi K, Babich D, Bangert JD, Kluiber 208.
R. Histologic evaluation of rat mammary 66. Zhou XD. Cryosurgery for liver cancer: experi-
tumor necrosis by interstitial Nd:YAG laser mental and clinical study (author's trans).
hyperthermia. Lasers Surg Med 1992;12(2): [Article in Chinese] Zhonghua Wai Ke Za Zhi
159-164. 1979;17(6):480-483.
54. Sakai T, Fujishima I, Sugiyama K, Ryu H, 67. Ravikumar TS, Kane R, Cady B, et al. Hepatic
Uemura K. Interstitial laserthermia in neuro- cryosurgery with intraoperative ultrasound mon-
surgery. J Clin Laser Med Surg 1992;10(1):37- itoring for metastatic colon carcinoma. Arch
40. Surg 1987;122:403.
55. Seki T, Kubota Y, Wakabayashi M, et al. Percu- 68. Onik G, Gilbert J, Hoddick W, et al. Sonographic
taneous transhepatic microwave coagulation monitoring of hepatic cryosurgery in an experi-
therapy for hepatocellular carcinoma proliferat- mental animal model. AJR 1985;144(5):1043-
ing in the bile duct. Dig Dis Sci 1994;39(3): 1047.
663-666. 69. Charnley RM, Dorn J, Morris DL. Cryotherapy
56. Seki T, Wakabayashi M, Nakagawa T, et al. Ultra- for liver metastases: a new approach. Br J Surg
sonically guided percutaneous microwave co- 1989;76(10):1040-1041.
agulation therapy for small hepatocellular 70. Onik G, Rubinsky B, Zemel R, et al. Ultrasound-
carcinoma. Cancer 1994;74(3):817-825. guided hepatic cryosurgery in the treatment of
57. Murakami R, Yoshimatsu S, Yamashita Y, metastatic colon carcinoma. Preliminary results.
Matsukawa T, Takahashi M, Sagara K. Treatment Cancer 1991;67(4):901-907.
of hepatocellular carcinoma: value of percuta- 71. Onik GM, Atkinson D, Zemel R, et al.
neous microwave coagulation. AJR 1995;164(5): Cryosurgery of liver cancer. Sernin Oncol 1993;
1159-1164. 9:309.
58. Hamazoe R, Hirooka Y, Ohtani S, Katoh T, 72. ter Harr G, Sinnett D, Rivens 1. High intensity
Kaibara N. Intraoperative microwave tissue focused ultrasound-a surgical technique for the
coagulation as treatment for patients with non- treatment of discrete liver tumours. Phys Med
resectable hepatocellular carcinoma. Cancer Bioi 1989;34(11):1743-1750.
1995;75(3):794-800. 73. Chapelon JY, Margonari J, Bouvier R, Cathignol
59. Dong B, Liang P, Yu X. US-guided microwave in D, Gorry F, Gelet A. [Tissue ablation by focused
the treatment of liver cancer: experimental study ultrasound] [Article in French]. Prog Urol
and preliminary clinical application. Zhonghua 1991;1(2):231-243.
Yi Xue Za Zhi 1996;76(2):87-91. 74. Prat F, Ponchon T, Berger F, Chapelon JY,
60. Sata M, Watanabe Y, Ueda S, et al. Two long-term Gagnon P, Cathignol D. Hepatic lesions in the
survivors after microwave coagulation therapy rabbit induced by acoustic cavitation. Gastro-
for hepatocellular carcinoma: a case report. enterology 1991;100(5 pt 1):1345-1350.
Hepatogastroenterology 1996;43(10):1035-1039. 75. Yang R, Reilly CR, Rescorla FJ, et al. High-
61. Yamanaka N, Tanaka T, Oriyama T, Furukawa K, intensity focused ultrasound in the treatment
Tanaka W, Okamoto E. Microwave coagu- of experimental liver cancer. Arch Surg 1991;
lonecrotic therapy for hepatocellular carcinoma. 126(8):1002-1009; discussion 1009-1010.
World J Surg 1996;20(8):1076-1081. 76. Yang R, Sanghvi NT, Rescorla FJ, et al. Extra-
62. Cahan WG. Cryosurgery of massive recurrent corporeal liver ablation using sonography-
cancer. Panminerva Med 1975;17(11-12):359- guided high-intensity focused ultrasound. Invest
361. Radiol 1992;27(10):796-803.
63. Dutta P, Montes M, Gage AA. Large volume 77. Yang R, Kopecky KK, Rescorla FJ, Galliani CA,
freezing in experimental hepatic cryosurgery. Wu EX, Grosfeld JL. Sonographic and com-
Avoidance of bleeding in hepatic freezing by an puted tomography characteristics of liver abla-
improvement in the technique. Cryobiology tion lesions induced by high-intensity focused
1979;16(1):50-55. ultrasound. Invest Radiol 1993;28(9):796-801.
16 J.p. McGahan and v.A. van Raalte

78. Sibille A, Prat F, Chapelon lY, et a1. Extracor- 80. Gelet A, Chapelon lY, Margonari 1, et a1.
poreal ablation of liver tissue by high-intensity Prostatic tissue destruction by high-intensity
focused ultrasound. Oncology 1992;50(5):375- focused ultrasound: experimentation on canine
379. prostate. 1 EndouroI1993;7(3):249-253.
79. Chapelon lY, Margonari 1, Theillere Y, et a1. 81. Gelet A, Chapelon lY, Margonari 1, et a1. High-
Effects of high-energy focused ultrasound on intensity focused ultrasound experimentation on
kidney tissue in the rat and the dog. Eur Urol human benign prostatic hypertrophy. Eur Urol
1992;22(2):147-152. 1993;23(suppl1):4~7.
2
Epidemiology: How to Appraise the
Ablation Literature Critically
Craig Earle

Example of an Abstract myocardial infarction, median progression-free


survival was 13 months. Tumor response was
PURPOSE: To evaluate the effectiveness of seen in 87% of cases. Median time to death or
ablation for hepatic tumors. METHODS: The last follow-up was 18 months: 16 months for
medical records of patients with either primary nonsurvivors, and 20 months for survivors.
or secondary hepatic tumors who underwent Complications occurred in 27% of patients and
ablative procedures from February 1991 to May included one skin burn, one postoperative hem-
2001 at a single institution were retrospectively orrhage from hepatic parenchyma cracking,
reviewed. One hundred nine patients with 140 and two hepatic abscesses. Only 4.7% locally re-
tumors ranging in size from 0.5 to 12 cm curred, although 37% have died of their cancer,
in diameter were treated. The diagnoses were and another 28% developed metastatic disease
colorectal cancer (n = 69), hepatoma (n = 15), at other sites. CONCLUSION: Ablation may be
ovarian cancer (n = 8), cholangiocarcinoma (n effective in allowing patients to undergo liver
= 4), carcinoid (n = 7), one each of leiomyosar- surgery and achieve better survival.
coma, testicular cancer, and endometrial cancer,
and other tumors (n = 3). Ablation was used to Clinical epidemiology spans the breadth of
treat 90 tumors: 47 percutaneously, 23 laparo- health and disease, from evaluating causation
scopically, and 20 intraoperatively. Additional and risk, to strategies for disease prevention,
tumors were identified by intraoperative ultra- to establishing diagnosis and prognosis, and to
sound in 37% of the patients taken to surgery evaluating interventions. As the focus of this
despite extensive preoperative imaging. In 45%, book is on techniques of tumor ablation, the
radiofrequency ablation (RFA) was combined focus here is on the evaluation of this form of
with resection or cryoablation or both. Alcohol intervention, in particular looking at pitfalls
ablation was performed on those patients who the reader may encounter when trying to read
were found to have residual tumor after the the ablation literature. The composite abstract
initial ablative procedure. Ten patients under- above was extracted from abstracts of pub-
went a second procedure and three had a third lished papers describing clinical studies of
for progressive or recurrent disease. Neoadju- tumor ablation. Although no single study was
vant chemotherapy was used in 19 cases, intra- this uninterpretable, it illustrates the challenges
hepatic treatment in 10, and postoperative a reader faces when trying to make sense of
chemotherapy was given to 33 patients. Follow- ablation studies. In the end, clinicians must be
up ranged between 12 and 28 months. able to take the results reported in a study and
RESULTS: If we exclude the six cases in which evaluate whether the intervention in question
it was clearly impossible to destroy the liver is likely to benefit the patient they have before
tumors and the one death due to postprocedure them.

17
18 C. Earle

When assessing a medical article, the reader a sense, is a form of historic controls. If the out-
must first judge whether the results are valid, comes of the study exceed these expectations,
and then try to fit them into the greater context the treatment can be judged to be promising;
of the field. A review conducted in June 2002 of however, the reader must be careful to question
clinical trials in adults reported in English in the at least three important components of the
past 10 years of the MEDLINE database using study: (1) Who were the patients? (2) What was
the intersection of key words "tumor ablation", the intervention? (3) What are the outcomes,
"radiofrequency ablation", or "cryoablation" and how are they assessed?
with "neoplasms", yielded 95 citations, of which
49 were relevant original studies. All but one Who Were the Patients?
was an uncontrolled case series. These ranged
in size from four to 308 patients. There were Selection Bias
large variations in reported outcomes, much of
The first concern about encouraging results
which likely is due to differing characteristics of
in a nonrandomized trial is usually that the
the patients, different ablative techniques, other
patients have been "cherry picked," meaning
interventions the patients received, or the way
that selection bias has influenced the results.
the outcomes were assessed, analyzed, or re-
Because no study can observe every patient in
ported. As a result, it is clear that understand-
the world with a given clinical problem, the
ing the ablation literature requires the ability to
patients in trials are a sample of the entire uni-
evaluate such noncomparative studies and to
verse of patients. It is critical to determine how
recognize their strengths and limitations.
representative this sample is of the population
of interest. For example, patients with better
Appraising Noncomparative performance status are known to tolerate inter-
ventions better, are more likely to have favor-
Studies able outcomes like response to treatment, and
tend to have longer survival than those who are
One of the main strengths of randomized con-
more ill. Patients with a lesser disease burden
trolled trials is that randomization provides
usually do better in these respects, while those
the appropriate control group for comparison
who have been heavily pretreated do worse.
of outcomes, as patients in the intervention and
Patients drawn entirely from academic referral
control arms should be similar in all ways
centers tend to be younger and more proactive
except for the random allocation to the differ-
and health conscious than those in the com-
ent treatment groups. If randomization is
munity. A study consisting only of patients with
carried out correctly in a study of sufficient
characteristics making them likely to have
sample size, both known and unknown prog-
good outcomes anyway, can almost guarantee
nostic features should be balanced between the
promising results. Taken to extremes, exclusion
intervention and control groups. Nonrandom-
of patients who are less likely to respond to
ized studies with a control group attempt to do
treatment can theoretically eventually lead to
the same thing, although there is always a risk
100% response rate. Authors should provide
that the control group is different from the
enough information for readers to know the
intervention group in some way that could
entry criteria and patient characteristics to
influence the results. For example, more robust
assess whether the sample is representative of
patients are generally more likely to have been
their patients.
selected to undergo aggressive therapy. Even
in studies without an explicit control group,
Assembly of the Inception Cohort
however, such as the bulk of the ablation liter-
ature, implicit comparison of outcomes is usu- An important safeguard against selection bias
ally being made with the reader's sense of what is a clear description of how the inception
the outcomes of such patients would have been cohort was assembled. This involves communi-
without the intervention being studied. This, in cating an understanding of the number of eligi-
2. Epidemiology 19

ble and ineligible patients seen within the study demonstrate real-world effectiveness, the study
period so that the reader can estimate the would be designed to take consecutive patients
degree of selection used to obtain the study with perhaps less strict entry criteria to include
population (1). The reader must be able to the breadth of patients to whom the interven-
follow the flow of patients including referral tion is likely to be applied, and carry out the
patterns, seeing that consecutive patients (not intervention outside of highly specialized set-
selected patients) were approached for the tings. While it may be more difficult to obtain a
study, noting how many of those refused and positive result in such a study, the generaliz-
how many were later excluded, and the reasons ability of these results makes them much more
for their exclusion. Ultimately, one should be compelling.
able to account for all patients who did or did
not participate, and know the denominator Excluded Patients
used for analyses at all times.
The fewer the patients who refuse to be
included or are excluded from analysis, the
Entry Criteria better. There are many examples from the
Another safeguard against selection bias is medical literature showing that patients who
to have rigorously defined entry criteria that agree to participate in clinical trials and who
ensure that the patient group is relatively are compliant with therapy tend to have better
homogeneous. For example, although it seems outcomes than those who do not. For example,
obvious, histologic confirmation of disease if patients who suffer complications from a pro-
should be required for entry into an ablation cedure or for whom the procedure is consid-
study. However, several relatively large series ered technically incomplete are excluded from
have not required this, leading at least one the analysis for being "inevaluable", the results
reviewer to wonder, "How many of the sur- are likely to be biased. The easiest way to
vivors ... may have had cryotherapy for lesions improve the results of a trial are to exclude the
that were not in fact metastases?" (2). This must patients who didn't tolerate or respond to treat-
be kept in mind when comparing outcomes to ment! Once again, patients with the best under-
those for surgical resection, as a resected tumor lying performance status and biology tend to
will always be pathologically confirmed, while be more likely to tolerate medical interven-
the same is not necessarily true of an ablated tions successfully. Thus, a study with very few
lesion. patients excluded is likely to provide more
The way a study sample is constructed valid results.
depends in part on whether the primary goal
of the study is to demonstrate internal validity
and efficacy (Can something work under ideal
What Was the Intervention?
conditions?) versus external validity and Ablative technologies are relatively new and
effectiveness (Does it work? Are the results are still undergoing development. As a result,
generalizable to the real world?). Selection of trials may not be comparable because of the
patients in clinical trials always involves trade- different techniques, instruments and manufac-
offs between these concepts. Ideally, to prove turers, and operator abilities. Furthermore, this
that an intervention works in a specific situa- continuous evolution means that the results of
tion, investigators would select patients who trials are often obsolete by the time they are
were very similar in all clinical and demo- published. Also, unless ablation is followed by
graphic characteristics. If the intervention is surgical resection, it is usually not possible to
demonstrated to work and the trial is done well, assess the adequacy of the ablation, unlike the
it is efficacious in that particular clinical situa- ability to pathologically assess surgical margins.
tion with those patients (internal validity). A related consideration is whether the tech-
However, it may be unclear whether the results nique is likely to be adequately performed
could be extrapolated to other scenarios. To when brought out of a highly specialized setting
20 C.Earle

into general use. Problems with translating a scans at prespecified intervals), the threshold
procedure into practice can greatly reduce the for measurements that were used (e.g., 50%
effectiveness of an efficacious intervention. decrease in the sum of the bidimensional
Study protocols also differ widely in whether areas of all tumors required to be deemed a
multiple lesions are ablated, the maximum size response), and whether the response had to be
of lesion to be attempted, and whether there is "durable", that is, maintained for a prespecified
co-intervention. For example, several studies period of time (e.g., on two computed tomog-
examined above allowed patients following raphy [CT] scans at least 2 months apart). This
ablation to subsequently receive chemother- is important because minor responses of short
apy. As a result, it is impossible to discern duration can occur entirely on the basis of
whether observed stable disease is primarily measurement error in diagnostic imaging alone
due to innate biology, ablation, or subsequent (3,4). In ablation studies, response is a difficult
systemic therapy. This heterogeneity in inter- end point because the ablation procedure often
vention makes much of the ablation literature results in radiographic abnormalities that make
difficult to interpret. it difficult to measure the tumor. Measuring
decreased uptake of a lesion on positron emis-
sion tomography (PET) scan has not been ade-
What Are the Outcomes, and How quately validated.
Were They Assessed?
The only outcomes that really matter to Time to Progression
patients are the quantity and quality of life
Time to progression often is evaluated in abla-
(primary end points). Anything else is a surro-
tion studies. However, it has little meaning
gate or intermediate end point that may corre-
unless it is of long duration. As Baar and
late to a greater or lesser extent with primary
Tannock (1) point out, any tumor with expo-
end points (Table 2.1).
nential growth and a volume doubling time of
2 months (an average value for human solid
Response
tumors) will require at least 1 month to increase
Response to therapy, particularly the control of its cross-sectional area by 25%. CT scans also
single lesions, has been shown many times to be may lack the sensitivity to determine when an
a poor surrogate for either survival or quality ablated liver metastasis has started to grow
of life. If response is used as an outcome, it must again until its size increases substantially. Last,
be clearly defined a priori in terms of the strin- the time to progression depends very much on
gency of criteria (e.g., evaluated on high-quality how often it is assessed. A study that does

TABLE 2.1. Considerations when evaluating the ablation literature.


1. Who were the patients? Is there evidence of selection bias?
a. How was the inception cohort assembled? What were the referral patterns? Were consecutive patients considered
for study? Is the sample size large enough?
b. Were there clearly defined entry criteria: disease type, histologic confirmation of lesions, restrictions on tumor size
and number? Are the patients representative of the population usually considered for this procedure?
c. Can you follow all patients approached and excluded to reach the final denominator?
2. What was the intervention?
a. Was the protocol specifically defined and uniformly followed?
b. How was the adequacy of ablation evaluated?
c. What co-interventions were permitted?
3. What were the outcomes, and how were they measured? Was there objective, preferably blinded assessment of
prespecified intermediate outcomes?
a. Was there a definition of how response to treatment would be defined, and how often it would be assessed?
b. Was there adequate follow-up time to evaluate survival?
c. Were toxicity and complications reported in detail?
2. Epidemiology 21

scans every 2 months is likely to have shorter ablations to ensure that patients have had an
times to progression than one that evaluates adequate time to manifest recurrent disease.
patients only every 3 months. Such studies Survival in the palliative setting is even more
cannot easily be compared. For these reasons, difficult to assess. Here it is very likely
the way outcomes are measured should be as that patients will undergo some form of co-
objective as possible, specified a priori, and, intervention because of the severity of their
when possible, evaluated blindly. For example, disease. It is also very difficult to compare
radiologists could be asked to evaluate lesions patients if they are in different places in their
out of chronologie order to get objective mea- disease course (e.g., newly diagnosed versus
sures of size. In this way, any optimism about heavily pretreated), making eligibility criteria
the effectiveness of treatment that could sys- an important consideration. Moreover, if
tematically affect the results of the study can be patients with more than one type of cancer were
minimized. included, they may have very different prog-
noses. Comorbid diseases and competing risks
also become important in this situation. Such
Survival
potentially confounding factors must be taken
Evaluation of survival can be divided into two into account in the analysis through stratifica-
general situations: those in which treatment is tion (comparing like patients only with like) or
potentially curative, and those in which it is pal- statistical adjustment. Analyses or survival by
liative only. In a situation in which the disease response ("patients who responded to treat-
is considered uniformly and predictably fatal ment lived longer") are usually not considered
without treatment, such as that of a solitary valid because the patients who respond are
liver metastasis from colon cancer, demonstra- usually those with better prognostic features as
tion of disease-free survival 5 years after an well.
intervention is a strong indication of efficacy.
However, there is a small proportion of patients
Quality of Life
with indolent disease who may survive at least
5 years without intervention. While such a There has been little research into the quality-
situation is more likely in prostate cancer of-life effects of tumor ablation. In potentially
than lung cancer, it has been described in both curative settings, if ablation can be shown to be
(5,6). Therefore, although the results may be as effective as surgery, it would likely avert much
provocative, any possibility of selection bias of the trauma and morbidity associated with
still must be evaluated. It is important to know many operative procedures. Most patients with
not only that a number of such patients exists, incurable metastatic disease have relatively
but what proportion of the population of inter- small tumors that are not causing specific symp-
est they represent. In this case, overall survival, toms such as pain. As a result, it is difficult to
including death from any cause, must be show that a procedure that usually causes at
reported. least some discomfort actually improves overall
Cause-specific survival, which evaluates only quality of life. Advanced cancer, however, is
whether patients died of recurrent or progres- associated with several constitutional symptoms
sive cancer, can be misleading, as judgment such as anorexia, weight loss, fatigue, and
is required in assigning the cause of death, anemia. These effects, mediated through
potentially leading to misclassification of the cytokines such as tumor necrosis factor (TNF),
outcome. Furthermore, even though the litera- correlate with overall body tumor burden. If
ture suggests that ablation generally is safer ablation could decrease tumor burden, it is pos-
than surgical procedures (2), any patients who sible that it could ameliorate some of these more
die from complications of the procedure should subtle cancer-related symptoms and improve
be included in survival analyses. Last, it is psychological well-being, thereby improving
crucial that adequate follow-up time be allowed quality of life. However, there have been no
to evaluate survival after potentially curative studies looking at these outcomes. Whether
22 C. Earle

delay or prevention of later progressive symp- ment protocols, and precise outcome measure-
toms provides overall benefit would need to be ments over adequate follow-up time. Attempts
evaluated in a randomized trial. to synthesize existing data are useful, but are
The toxicity of treatment should be described prone to publication bias: series with poor
explicitly in reports of tumor ablation results. results are less likely to be published, and if
Any serious event, even if it could have been published, are usually in less prominent jour-
due to the underlying disease or an unrelated nals. If warranted, based on such studies,
cause, should be reported in detail. Over time, randomized trials will be needed in most
and with randomized studies, it will become applications before ablation can rightly take its
apparent whether these are truly attributable to place among the other standard treatments for
the procedure or not. Subjective statements, cancer. As promising as these techniques are,
such as "the treatment was generally well tol- we must ensure that they do not become
erated", are not useful and may have very dif- entrenched in medical practice before they are
ferent meanings for different readers. studied rigorously, as this can make random-
ization in trials very difficult. The history of
Statistical Considerations medicine tells us that we have been wrong
before about interventions that seem as if they
Regarding the analysis of results, sample size is
should work. Therefore, it is imperative that the
the most important factor for determining the
benefits be proven before subjecting large
precision of results. For example, a positive
numbers of patients to the risks and expense of
outcome for six patients in a study of 10 patients
ablation. At that time, added considerations
is interesting, but much less certain than a good
such as cost-effectiveness and health policy will
result for 60 patients out of 100. Statistically,
need to be addressed as well.
this manifests by being able to draw more
narrow confidence limits around an estimate of
response rate derived from the larger study.
Chance, or random variation, is less likely to References
cause the point estimate to be different from the
1. Baar J, Tannock IF. Analyzing the same data in
true underlying response rate for the entire pop- two ways: a demonstration model to illustrate the
ulation of patients in a larger study. reporting and misreporting of clinical trials. J Clin
OncoI1989;7:969-978.
Conclusion 2. Tandan VR, Harmantas A, Gallinger S. Long-term
survival after hepatic cryosurgery versus surgical
resection for metastatic colorectal carcinoma: a
Techniques for tumor ablation are promising critical review of the literature. Canadian Journal
and becoming more refined with time. However, of Surgery 1997;40:175-181.
the current literature is dominated by small case 3. Moertel CG, Hanley JA. The effect of measur-
series that are difficult to interpret and compare ing error on the results of therapeutic trials in
with each other. Evaluation of different ablative advanced cancer. Cancer 1976;38:388-394.
procedures through comparison of nonrandom- 4. Warr D, McKinney S, Tannock IF. Influence of
ized studies requires, in addition to assessment measurement error on assessment of response to
of the quality of the study, synthesis of whether anticancer chemotherapy: Proposal for new crite-
the patients undergoing the procedures were ria of tumor response. J Clin Oncol 1984;2:1040-
representative, whether the intervention was 1046.
5. Marcus PM, Bergstralh EJ, Fagerstrom RM, et al.
applied correctly, and whether the important
Lung cancer mortality in the Mayo Lung Project:
outcomes were measured in a valid way. For the impact of extended follow-up. J Natl Cancer Inst
reader, all of this is also dependent on the quality 2000;92:1308-1316.
of the reporting of the trial. 6. Holmberg L, Bill-Axelson A, Helgesen F, et aI. A
The role of ablation in almost all clinical sit- randomized trial comparing radical prostatec-
uations needs more study in large prospective tomy with watchful waiting in early prostate
trials with rigorous entry criteria, uniform treat- cancer. N Eng J Med 2002;347:781-789.
3
Image-Guided Tumor Ablation:
Basic Science
Muneeb Ahmed and S. Nahum Goldberg

Minimally invasive strategies in image-guided need to be modified for the increasing use of
tumor ablation are gaining increasing attention ablation to treat tumors in such locations as the
as viable therapeutic options for focal primary kidney (7,8), lung (9,10), bone (11), and breast
and secondary hepatic malignancies (1–3). (12).
Although liver transplantation continues to be Current percutaneous tumor ablation
the standard for cure of hepatocellular carci- techniques can be divided into two broad
noma (HCC), there remains a clear need for categories, chemical and thermal, based on dif-
treatment alternatives in the large population ferences in mechanisms of inflicting tissue
of HCC patients unable to qualify for liver injury. Chemical ablation strategies involve the
transplantation surgery (4). For hepatic metas- percutaneous intratumoral administration
tases, while conventional surgical resection has (or instillation) of ablative substances, such as
demonstrated acceptable rates of success (5) in ethanol and acetic acid. Thermal ablation
carefully selected patient populations, several strategies utilize alterations in tissue tempera-
classes of minimally invasive, image-guided ture to induce cellular disruption and tissue
therapeutic strategies are being vigorously coagulation necrosis. Potential strategies within
explored as practical alternatives (2,3). Possible this latter group include ablation via thermal
advantages of minimally invasive therapies energy (radiofrequency, microwave, laser, ultra-
compared to surgical resection include the sound) and freezing via cryotherapy. Given that
anticipated reduction in morbidity and mortal- a wide range of technologies is being applied,
ity, lower cost, the ability to perform procedures this chapter provides a conceptual framework
on outpatients, and the potential application in for the principles and theories that underlie
a wider spectrum of patients, including nonsur- focal tumor ablation therapies. Furthermore,
gical candidates. The role of these strategies in the basic principles and tissue–therapy interac-
the HCC patient population is further ampli- tions of widely accepted modalities are
fied by the presence of severe underlying reviewed. Finally, current and future directions
hepatic dysfunction and coagulopathy in many of research also will be discussed.
patients, which complicates or obviates poten-
tial surgery; the absence of adequate alternative Overview: Theory of
therapeutic options; and the favorable initial
equivalent treatment response of HCC com- Image-Guided Tumor Ablation
pared to surgical resection (2,6). Furthermore,
Principles of Minimally Invasive Focal
the availability of various techniques allows
Tumor Ablation
treatment tailored to patient-specific disease,
which is anticipated to further increase long- The ultimate strategy of image-guided tumor
term success rates. These techniques will also ablation therapies encompasses two specific

23
24 M. Ahmed and S.N. Goldberg

objectives: First and foremost, all tumor abla- for the longest period of all image-guided
tion therapies, through applications of energy ablative therapies, with the largest number of
or chemical substances, attempt to completely studies reporting long-term follow-up. Injected
eradicate all viable malignant cells within a des- alcohol achieves its effect on tumor cells by two
ignated area. Based on earlier studies examin- primary mechanisms: (a) as it diffuses into neo-
ing tumor progression for patients undergoing plastic cells, alcohol results in immediate dehy-
surgical resection, and the demonstration of dration of the cytoplasm, protein denaturation,
the presence of viable malignant cells beyond and consequent coagulation necrosis; and (b)
visible tumor boundaries, tumor ablation ther- alcohol entering the local circulation leads to
apies also attempt to include a l.Ocm "ablative" necrosis of the vascular endothelium and
margin of seemingly normal tissue that is subsequent platelet aggregation, that results in
presumed to also contain viable malignant cells vascular thrombosis and ultimately ischemic
(1). Second, while complete tumor eradication tissue necrosis (15,16).
is of primary importance, specificity and accu- The treatment of primary HCC with ethanol
racy of therapy also are required. One of the injection has been considerably more success-
significant advantages of these therapies over ful than the treatment of liver metastases
conventional standard surgical resection is the because of tumor characteristics, including
potential minimal amount of normal tissue loss softer tumor composition and a delineated
that occurs. For example, in primary liver capsule surrounded by cirrhotic liver that
tumors, in which functional hepatic reserve is a results in limited diffusion and increased
primary predictive factor in long-term patient concentration within the target (17). Hepatic
survival outcomes, image-guided tumor abla- tumors can be treated with ethanol ablation
tion therapies have documented success in using either multiple treatment sessions (1), or
minimizing iatrogenic damage to cirrhotic a single session in which a large volume of
parenchyma surrounding focal malignancies. alcohol is injected, usually via many separate
Therefore, one of the objectives of tumor abla- needle punctures (18). However, this latter
tion therapies is maintaining a balance between approach has been largely abandoned in favor
adequate tumor destruction and minimal of thermal therapies, as it is performed only
damage to normal tissue and surrounding under general anesthesia, and has higher
structures. reported complication rates. Furthermore,
studies have demonstrated that the efficacy of
percutaneous chemical ablation with ethanol is
Chemical Ablation variable and significantly influenced by tumor
type. In cases of secondary hepatic malignan-
Intratumoral administration of chemically cies, the heterogeneous and dense fibrous
ablative substances, such as ethanol and acetic nature of the tumor tissue limits the diffusion
acid, has documented efficacy as a method for of ethanol within the tumor (19). Efficacy rates
percutaneous focal tumor destruction. Ethanol of ethanol instillation for hepatic metastases
instillation has had widely reported success are poor, and therefore ethanol is not recom-
in the treatment of focal hepatocellular carci- mended for the treatment of such malignancies.
noma. More recently, several investigators also
have documented similar or greater efficacy
with intratumoral instillation of acetic acid.
Acetic Acid
Intratumoral instillation of acetic acid recently
has received increased attention as another
Ethanol
chemical substance with documented efficacy
Ethanol ablation, classically known as percuta- in percutaneous chemical tumor ablation.
neous ethanol instillation (PEl), is used princi- Given several limitations of ethanol instillation,
pally to treat HCC in patients with cirrhosis including poor and uneven distribution, and
(13,14). Ethanol ablation has been utilized limited success in a wider range of tumor
3. Image-Guided Tumor Ablation: Basic Science 25

types, several studies have investigated the agent no longer is used. In the neck, chest,
use of acetic acid as a viable alternative to abdomen/pelvis, and extremities, cryoablation
ethanol for cases undergoing chemical ablation is performed using a closed cryoprobe that is
(20,21). The mechanisms of tissue injury, in- placed on or inside a tumor. The two main types
cluding direct cellular damage through the of systems use either gas or liquid nitrogen, or
induction of intracellular dehydration and vas- argon gas. Temperatures are measured either at
cular endothelial injury and resultant thrombo- the tip of the cryoprobe or in the handle.
sis, are similar for both acetic acid and ethanol Although in the past, cryoablation needed to
(22). However, in preliminary animal studies, be performed in the setting of an open pro-
Ohnishi et al (22) have demonstrated signifi- cedure, technologic developments have led
cantly greater coagulation necrosis diameters to smaller probes that can be placed per-
for acetic acid solutions with concentrations cutaneously under magnetic resonance imag-
above 20%, compared to 100% ethanol. This ing (MRI) or computed tomography (CT)
suggests that the diffusion ability of acetic acid guidance (29).
solutions may exceed those of ethanol, likely Several mechanisms have been identified
based on the effects of acetic acid on dissolving through which cryoablation induces tissue
fibrous interstitial tissue; this factor may poten- injury (30). First, cryoablation induces direct
tially result in improved tumor ablation efficacy cellular injury through tissue cooling. At low
compared to ethanol (23). However, further cooling rates, freezing primarily propagates
research is required to fully characterize and extracellularly, which draws water from the cell
understand the interactions of acetic acid with and results in osmotic dehydration (31). The
tumor tissue. intracellular high solute concentration that
develops leads to damage to enzymatic systems
and proteins, and injury to the cellular mem-
brane (32,33). At faster cooling rates, water is
Thermal Ablation Therapies trapped within the cell, intracellular ice forma-
tion occurs, and organelle and membrane injury
Thermal ablation strategies attempt to destroy
results (31,34). A second hypothesized mecha-
tumor tissue in a minimally invasive manner
nism of cryoablative tissue destruction is that
by increasing or decreasing temperatures suffi-
of vascular injury, manifested as mechanical
ciently to induce irreversible cellular injury.
injury to the vessel wall, direct cellular injury
These strategies can be broadly divided into
to various cells lining the vessel, and post-thaw
two groups, based on whether they utilize tissue
injury from reperfusion (30). Mechanical injury
freezing or heating: cryoablation or hyperther-
to the vessel wall from intravascular ice forma-
mic ablation therapies.
tion results in increased vessel leakiness,
reduced plasma oncotic pressure, and perivas-
cular edema (35). Endothelial injury further
Cryoablation
results in exposure of underlying connective
Cryoablation, used successfully in the focal tissue and subsequent thrombus formation
treatment of several types of tumors, achieves (36). Damage to endothelial cells occurs in a
focal tissue destruction by alternating between fashion similar to the direct cellular injury
sessions of freezing followed by tissue thawing described above. Reperfusion injury results
(24-28). These reductions in tissue tempera- from the release of vasoactive factors after the
tures are achieved through specially con- tissue thaws, which leads to vasodilation and
structed probes that are placed within the increased blood flow into treated areas. Subse-
target tissue. Liquid forms of inert gases are quent high oxygen delivery (37) and neutrophil
then cycled through the hollow probes, result- migration (38) into the damaged area result in
ing in tissue cooling. In the past, liquid nitrogen increased free radical formation, and ulti-
was placed directly on tissue, but with the mately, through peroxidation of membrane
exception of dermatologic applications, this lipids, further endothelial damage.
26 M. Ahmed and S.N. Goldberg

The degree to which each of the above mech- energy sources have been used to provide the
anisms of cellular and tissue injury determine heat necessary to induce coagulation necrosis.
overall tissue destruction is governed by Focal high temperature (>50°C) hyperthermic
characteristics of the administration algorithm, ablation therapies use microwave, radiofre-
that include freezing and thawing rate, the nadir quency (RF), laser, or ultrasound energies to
(i.e., coldest) temperature reached, and the generate isolated increases in tissue tempera-
duration that the temperature is maintained ture. This focal heating primarily is achieved
(30). At low and high tissue cooling rates, through the placement of applicators in the
osmotic dehydration and intracellular ice for- center of the target, around which heating
mation, respectively, induce direct cellular occurs. This approach is similar for all thermal
injury, while cooling rates in between these ablation strategies, regardless of the type of
extremes do not inflict as much tissue injury energy source used. Currently, the greatest
(31). Maintaining freezing temperatures for number of both clinical and experimental
longer durations also may increase the amount studies have been performed using RF-based
of damage by facilitating recrystalization, ablative devices. Therefore, our review of basic
whereby smaller ice crystals coalesce to create principles of focal high-temperature ablative
larger and more damaging crystals to reduce therapies primarily uses RF systems as a repre-
overall surface area and minimize free energy sentative model to describe the basic principles
(39). Similarly, thawing rate maximizes the time of focal thermal ablation, followed by brief
spent at lower temperatures. The effect of nadir discussions of the principles of heat generation
freezing temperature also determines the for other energy sources.
ability of cryoablative therapies to effectively
destroy tumor tissue. Studies on the effects of
freezing temperatures have demonstrated that
minimum lethal temperature is both variable
Radiofrequency
and cell-type specific (30).
While the algorithmic parameters detailed
Induction of Coagulation Necrosis
above influence overall tissue destruction, clin- Thermal strategies for ablation attempt to
ical outcomes are highly variable (30). Rates of destroy tumor tissue in a minimally invasive
tissue cooling and thawing vary and are non- manner, while limiting injury to nearby struc-
linear throughout treated tissue. Furthermore, tures (1,2,40,41). Treatment also includes a 5- to
tissue inhomogeneities, including differences in 10-mm ablative margin of normal tissue, based
vascularity, make it difficult to achieve similar on the uncertainty concerning the exact tumor
cooling characteristics throughout the tissue. margin and the possibility of potential micro-
Several biologic and physical characteristics scopic disease in the rim of tissue immediately
of the cell and surrounding microenvironment surrounding visible tumor (1,40). Cosman et al
also influence the degree of injury that occurs. (42) have shown that the resistive heating pro-
These include oxygen tension and metabolites duced by RF ablation techniques leads to heat-
in the tissue, cell cycle stage, state of membrane based cellular death via thermal coagulation
proteins before and after freezing, and the necrosis. Therefore, generated temperatures,
presence of minerals and proteins inside the and their pattern of distribution within treated
bloodstream. tissues, determine the amount of tumor
destruction.
Prior work has shown that cellular homeo-
High-Temperature Ablation
static mechanisms can accommodate slight
Tumor cells also can be destroyed effectively by increases in temperature (to 40°C). Although
cytotoxic heat from different sources. As long as increased susceptibility to damage by other
adequate heat can be generated throughout the mechanisms (radiation, chemotherapy) is seen
tumor volume, it is possible to accomplish the at hyperthermic temperatures between 42° and
objective of eradicating the tumor. Multiple 45°C, cell function and tumor growth continue
3. Image-Guided Tumor Ablation: Basic Science 27

even after prolonged exposure (43,44). Irre- This complex equation was further simplified to
versible cellular injury occurs when cells are a first approximation by Goldberg et al (2) to
heated to 46°C for 60 minutes, and occurs more describe the basic relationship guiding thermal
rapidly as temperature rises (45). The basis for ablation induced coagulation necrosis as, "coag-
immediate cellular damage centers on protein ulation necrosis = energy deposited x local tissue
coagulation of cytosolic and mitochondrial interactions - heat loss". Based on this, several
enzymes, and nucleic acid-histone protein com- strategies have been pursued to increase the
plexes (4~8). Damage triggers cellular death amount of coagulation necrosis by improving
over the course of several days. Coagulation tissue-energy interactions during thermal
necrosis is the term used to describe this ablation. Discussed below, these strategies have
thermal damage, even though ultimate mani- centered on altering one of the three parame-
festations of cell death may not fulfill strict ters of the simplified bioheat equation, either
histopathologic criteria of coagulative necrosis. by increasing RF energy deposition, or by mod-
This has significant implications with regard to ulating tissue interactions or blood flow.
clinical practice, as percutaneous biopsy with
histopathologic interpretation may not be a
reliable measure of adequate ablation. Optimal
RF Techniques to Improve
desired temperatures for ablation range from
Potential Outcome
50° to lOO°e. Extremely high temperatures Complete and adequate destruction by RF
(>lO5°C) result in tissue vaporization, which in ablation requires that an entire tumor be sub-
turn impedes the flow of current and restricts jected to cytotoxic temperatures. The studies
total energy deposition (49). described in the previous section demonstrated
that early use of RF as a strategy for percuta-
neous thermal ablation was inadequate for
Principles of the Bioheat Equation tumors larger than 1.8cm (49). Of the strategies
Success of thermal ablative strategies, whether to improve results of RF ablation, enhancement
the source is RF, microwave, laser, or high- of the electrode design has played an essential
intensity focused ultrasound, is contingent on role in achieving acceptable tissue tumor
adequate heat delivery. The ability to heat large coagulation.
volumes of tissue in different environments is
dependent on several factors that include both Modification of Probe Design
RF delivery and local physiologic tissue char-
Multiprobe Arrays
acteristics. Pennes (50) first described the rela-
tionship between this set of parameters as the Although lengthening the RF probe tip
bioheat equation: increased the volume of coagulation necrosis,
the cylindrical lesion shape does not corre-
spond well with the spherical geometry of most
PtctdT(r,t)/dt=V(ktVT)-Cb Pb mpt(T-Tb) tumors. In an attempt to address this inade-
+Qp(r, t) +Qrn (r, t) quacy, several groups tried to insert multiple
RF electrodes into tissue to increase heating in
a more uniform manner (51,52). However, for
where: complete ablation, this requires mUltiple over-
lapping treatments in a contiguous fashion to
PI> Pb = density of tissue, blood (kg/m3 ) treat tumors adequately. Hence, the time and
Cl> Cb = specific heat of tissue, blood (W s/kg C) effort required for this strategy make it imprac-
kt = thermal conductivity of tissue tical for clinical use.
m = perfusion (blood flow rate/unit mass Subsequent work centered on using several
tissue) (m 3/kgs) conventional monopolar RF electrodes utilized
Qp = power absorbed/unit volume of tissue simultaneously in an array, to determine the
Qm = metabolic heating/unit volume of tissues effect of probe spacing, configuration, and RF
28 M. Ahmed and S.N. Goldberg

application method (53). Spacing no greater liver to induce necrosis of up to 4.0 cm in the
than 1.5 cm between individual probes pro- long axis diameter, but could only achieve 1.4cm
duced uniform and reproducible tissue coagu- of necrosis in the short diameter. Although this
lation, with simultaneous application of RF system results in an overall increase in coagula-
energy that produced more necrosis than tion volume, the shape of necrosis is unsuitable
sequential application. Coagulation shape was for actual tumors, making the gains in coagula-
further dependent on spacing and the number tion less clinically significant.
of probes, and most often corresponded to the Desinger et al (60) described another bipolar
array shape. This improvement in RF applica- array that contains both the active and return
tion technique results in a significant increase electrodes on the same 2mm-diameter probe.
in the volume of inducible tissue necrosis, aug- This arrangement of probes eliminates the need
menting coagulation volume by up to 800% for surface grounding pads and the risk of
over RF application with a single conventional grounding pad burns (see below). Although
probe with similar tip exposure. there has been limited clinical experience with
Working to overcome the technical chal- this device, studies report coagulation of up to
lenges of multiprobe application, multi-tined 3 cm in ex vivo liver. Last, Lee's group (61,62)
expandable RF electrodes have been devel- reports using two multi-tined electrodes as
oped. These systems, produced by two com- active and return, to increase coagulation
mercial vendors, involve the deployment of a during bipolar RF ablation, and a switching
varying number of multiple thin, curved tines in technique between electrodes to increase abla-
the shape of an umbrella or more complex tion efficiency.
geometries from a central cannula (54,55). This
surmounts earlier difficulties by allowing place-
Internally Cooled Electrodes
ment of multiple probes to create large, repro-
ducible volumes of necrosis. LeVeen (56), using One of the limitations to greater RF energy
a 12-hook array, was able to produce lesions deposition has been overheating that surrounds
measuring up to 3.5 cm in diameter in in vivo the active electrode and leads to tissue char-
porcine liver by administering increasing ring, rising impedance, and RF circuit inter-
amounts of RF energy from a 50W RF gener- ruption. To address this, internally cooled
ator for 10 minutes. More recently, high-power electrodes have been developed that are
systems (up to 250W) have been developed capable of greater coagulation compared to
with complex, multi-tine electrode geometries. conventional monopolar RF electrodes. These
With these, preliminary reports claim coagula- electrodes contain two hollow lumina that
tion up to 3.5 cm in diameter (57,58). Further- permit continuous internal cooling of the tip
more, with the injection of adjuvant NaCI with a chilled perfusate, and the removal of
solution into tissue surrounding the RF elec- warmed effluent to a collection unit outside of
trode, increases in achievable coagulation up to the body. This reduces heating directly around
5 to 7cm in diameter now have been reported. the electrode, tissue charring, and rising imped-
ance, allowing greater RF energy deposition.
Goldberg et al (63) studied this modification
Bipolar Arrays
using an 18-gauge electrode. With internal elec-
Several groups have worked with bipolar arrays, trode tip cooling (of 15 ± 2°C using O°C saline
compared to the conventional monopolar perfusate), RF energy deposited into tissue
system to increase the volume of coagulation and resultant coagulation necrosis were signifi-
created by RF application. In these systems, cantly greater (p < .001) than that achieved
applied RF current runs from an active elec- without electrode cooling. With this degree of
trode to a second grounding electrode in place cooling, greater RF current could be applied
of a grounding pad. Heat is generated around without observing tissue charring. Maximum
both electrodes, creating elliptical lesions. diameters of coagulation measured 2.5, 3.0,4.5,
McGahan et al (59) used this method in exvivo and 4.4cm for the 1-,2-,3-, and 4cm electrode
3. Image-Guided Tumor Ablation: Basic Science 29

tips, respectively. With tip temperatures of 25° for 45 minutes to a single internally cooled elec-
to 35°C (less cooling), tissue charring occurred trode resulted in only 2.7 cm of coagulation
with reduced RF application, and resulting (p < .01).
necrosis decreased from 4.5 to 3.0cm in diam- Radiofrequency applied for 12 minutes to
eter. Similar results have been reported by electrode clusters produced 3.1 cm of coagu-
Lorentzen (64), who applied RF to ex vivo calf lation in in vivo liver, and 7.3 cm in in vivo
liver using a 14-gauge internally cooled elec- muscle. This was significantly greater than the
trode and 20°C perfusate. 1.8- and 4.3 cm sized lesions in in vivo liver and
Subsequent in vivo studies were performed muscle, respectively, that was seen with the use
in normal porcine liver and muscle tissue. of single internally cooled electrodes with
Within muscle tissue, lesion diameter increased similar technique (p < .01). When RF current
from 1.8 to 5Acm for 12 minutes of RF. Never- (2000mA) was applied for 30 minutes in in vivo
theless, in normal liver only 2.3 cm of coagula- muscle, 9.5 cm of coagulation was observed,
tion was observed, regardless of current applied including extension into the retroperitoneum
or treatment duration (to 30 minutes). Thus, in and overlying tissues.
vivo necrosis was significantly less than that In the initial clinical use of this clustered elec-
observed for ex vivo liver using identical RF trode device, 10 patients with large solitary
parameters (p < .01). In clinical studies, Solbiati intrahepatic colorectal metastases (4.2-7.0 cm)
et al (65) demonstrated 2.8 ± OAcm of coagu- were treated with a single RF application
lation for liver metastases. (12-15 minutes, 1600-1950mA) (66). Contrast-
enhanced CT postablation showed induced
coagulation necrosis that measured 5.3 ± 0.6 cm,
Cluster RF
with a minimum short axis diameter of 4.2 cm.
Based on success in inducing greater volumes The overall shape of coagulation was not spher-
of necrosis by using both multiprobe arrays and ical in several cases, and generally conformed to
cooling, initial experiments were performed to the tumor shape. In two cases, large blood
study the use of internally cooled electrodes in vessels limited the size of induced coagulation.
an array (66). Three 2cm tip exposure inter- The use of the internally cooled cluster elec-
nally cooled electrodes were placed at varying trode array offers the potential of large-volume
distances in ex vivo liver for simultaneous RF coagulation for clinical tumor ablation com-
application (llOOmA for 10 minutes). At pared to conventional monopolar RF elec-
spacing distances of 0.5 to 1.0cm apart, a trodes or single internally cooled electrodes.
uniform circular cross-sectional area of coagu- Spacing distances of less than 1 cm between
lation necrosis measuring 4.1 cm in diameter individual electrodes produces more spherical
was achieved. In contrast, electrode spacing of zones of necrosis with no intervening viable
1.5 to 2.5 em resulted in cloverleafed lesions tissue, compared to greater spacing distances.
with areas of viable tissue within the treatment While these results appear counterintuitive,
zone. postulating that these electrodes function as a
Using an optimized 0.5 cm spacing distance single large electrode can explain this phenom-
between probes, Goldberg et al (66) proceeded enon of greater coagulation necrosis with rela-
to study variations in electrode tip length and tively closely spaced electrodes.
duration of application on coagulation volume.
Tip lengths of 1.5 to 3.0cm were used in ex vivo
Pulsed RF Application
liver and in vivo porcine liver and muscle
systems for varied RF application (1400-2150 Pulsing of energy is another strategy that has
rnA) and duration (5-60 minutes). In ex vivo been used with RF and other energy sources
liver, simultaneous RF application to internally such as laser to increase the mean intensity of
cooled electrode clusters for 15, 30, and 45 energy deposition. When pulsing is used,
minutes produced 4.7,6.2, and 7.0cm of coagu- periods of high-energy deposition are rapidly
lation, respectively. In contrast, RF application alternated with periods of low-energy deposi-
30 M. Ahmed and S.N. Goldberg

tion. If a proper balance between high- and using continuous infusion of normal saline at
low-energy deposition is achieved, preferential 1 mL/min in experimental animal models and
tissue cooling occurs adjacent to the electrode human liver tumors. Miao et al (70,71) used a
during periods of minimal energy deposition novel "cooled-wet" technique to significantly
without significantly decreasing heating deeper increase RF-induced coagulation in both ex
in the tissue. Thus, even greater energy can be vivo and in vivo tissue using a continuous saline
applied during periods of high-energy deposi- infusion combined with an expandable elec-
tion, thereby enabling deeper heat penetration trode system. Kettenbach et al (72) have
and greater tissue coagulation (66). Synergy reported clinical use of these devices in 26
between a combination of both internal cooling patients. In these studies, the mechanisms
and pulsing has resulted in greater coagulation responsible for the increase in coagulation were
necrosis and tumor destruction than either not well characterized. This strategy seemed to
method alone (66). confirm an initial hypothesis that high local ion
Clearly, optimization is required for each concentration from NaCI injection could
system. Currently, at least one manufacturer increase the extent of coagulation necrosis by
has incorporated a pulsing algorithm into its effectively increasing the area of the active
generator design (67). An optimal algorithm surface electrode. Other possible explanations
with a variable peak current for a specified for the increase in coagulation include reduced
minimum duration was designed. Maximum effects of tissue vaporization (i.e., allowing
coagulation, which measured 4.5 cm in diame- for the probe to contact tissue despite the
ter, was achieved using initial currents greater formation of electrically insulating gases), or
than IS00mA, a minimum RF duration of 10 improved thermal conduction caused by diffu-
seconds of maximum deliverable current, with sion of boiling solution into the tissues.
a IS-second reduction in current to 100 rnA fol-
lowing impedance rises. This latter algorithm
produced 3.7 ± 0.6cm of necrosis in in vivo
Overcoming Physiologic Limitations
liver, compared to 2.9cm without pulsing (p < While modifications in electrode design and
.05). Remote thermometry further demon- application protocols have yielded increases in
strated more rapid temperature increases and coagulation volume compared to conventional
higher tissue temperatures when pulsed-RF monopolar RF systems, further gains through
techniques were used. Ramping of RF deposi- RF equipment modifications have not pro-
tion based on impedance also is used with some duced equivalent increases in clinical settings.
multi-tined expandable electrodes. Thus, an This is largely due to multiple and often tissue-
optimized algorithm for pulsed-RF deposition specific limitations that prevent heating of the
can increase coagulation over other pulsed and entire tumor volume (41,49). Most important is
conventional RF ablation strategies. the heterogeneity of heat deposition through-
out a given lesion to be treated. Recent atten-
tion has centered on altering underlying tumor
Perfused Electrodes
physiology as a means to advance RF thermal
Perfusion electrodes also have been developed ablation. Based on the simplified bioheat equa-
that have small apertures at the active tip to tion described earlier, these efforts can be
allow fluids (i.e., normal or hypertonic saline) divided into two broad categories. One exam-
to be infused or injected into the tissue before, ines altering local tissue interactions to permit
during, or after the ablation procedure. Curley greater RF energy input by changing tissue
and Hamilton (68) infused up to 10mL/min conductivity or increasing local heat conduc-
of normal saline in ex vivo liver for 4 minutes tion, while the other explores reducing heat loss
during RF application and achieved coagula- by modulating blood flow within treatment
tion that measured 2.6cm in diameter. zones. As will be shown, certain modifications
Similarly, Livraghi et al (69) have reported can have several simultaneous effects. For
coagulation of up to 4.1 cm in diameter example, saline a,nd iron compounds have been
3. Image-Guided Tumor Ablation: Basic Science 31

useful for improving energy deposition (73), et al (70,71) reported significant increases in
while continuous injection of saline during coagulation with normal saline infusion during
ablation has facilitated the spread of heat from RF ablation in ex vivo and invivo tissue.
the RF electrode into deeper tissues (70). Mod- Saline can have multiple effects, and in rele-
ulation of blood flow during RF application vant studies the mechanisms responsible for
can improve ablation by limiting perfusion- the increase in coagulation were not well
mediated tissue cooling (74). characterized.
More recent study has centered on exploring
Local Tissue Interactions the effects of volume and concentration of
NaCI on the RF coagulation volume. In normal
The effect of local tissue characteristics is porcine liver and agar tissue phantom models,
defined by thermal and electrical conductivity Goldberg et al (73) have demonstrated that
of surrounding tissue. Altering both of these, both NaCI concentration and volume influ-
either separately or simultaneously, can alter enced RF coagulation. These experiments
the pattern of thermal distribution with treated demonstrated that increased NaCl concentra-
tissue. tion increases electrical conductivity (which is
inversely proportional to the measured imped-
Improved Tissue Heat Conduction ance) and enables greater energy deposition in
As discussed for perfusion electrodes, im- tissues without inducing deleterious high tem-
proved heat conduction within the tissues by peratures at the electrode surface. However,
injection of saline and other compounds has this effect is nonlinear with markedly increased
been proposed (68,70,71). The heated liquid tissue conductivity that decreased tissue
spreads thermal energy farther and faster than heating. Under many circumstances (i.e., in
heat conduction in normal "solid" tissue. An the range of normal tissue conductivities) the
additional potential benefit of simultaneous increased conductivity can be beneficial for
saline injection is increased tissue ionicity that RF ablation in that it enables increased
enables greater current flow. Similarly, amplifi- energy deposition that increases tissue heating.
cation of current shifts with iron compounds, However, given less intrinsic electrical resis-
injected or deposited in the tissues prior to tance, increased tissue conductivity also in-
ablation, has been used for RF and microwave. creases the energy required to heat a given
Increased ferric ions, in the form of high doses volume of tissue. When this amount of energy
of supraparamagnetic iron oxide MRI contrast cannot be delivered (i.e., it is beyond the
agent, can raise the temperature of polyacryl- maximum generator output), the slope is nega-
amide phantoms during RF ablation (75). tive and less tissue heating (and coagulation)
will result. Thus, to achieve clinical benefit
(i.e., an increase in RF-induced coagulation),
Altered Tissue Thermal and
optimal parameters for NaCI injection need to
Electrical Conductivity
be determined both for each type of RF appa-
For a given RF current, the power deposition at ratus used and for the different tumor types and
each point in space is strongly dependent on the tissues to be treated.
local electrical conductivity. Several investiga- In an attempt to optimize the effects of
tors have demonstrated the ability to increase altered conductivity using injectable therapies,
coagulation volume by altering electrical con- Goldberg et al (73) initially investigated the
ductivity in tissues through saline injection effect of injecting different volumes of high-
prior to or during RF ablation. In both experi- concentration NaCI on the extent of RF-
mental animal models and human liver tumors, induced coagulation. Using an iterative,
Livraghi et al (60) achieved 4.1 cm of coagula- nonlinear simplex optimization strategy, param-
tion when continuous infusion of normal saline eters for NaCl injection were studied in in
at 1ml/min was administered during RF appli- vivo porcine liver over varying concentrations
cation. Additionally, Curley et al (68) and Miao (0.9-38%) and injection volume (0-25 mL)
32 M. Ahmed and S.N. Goldberg

using internally cooled electrodes following of hepatic vasculature in the vicinity of the
pulsed-RF algorithms for 12 minutes. Signifi- ablation. This reduced RF coagulation necrosis
cant increases in generator output, tissue heat- in in vivo settings is most likely a result of both
ing, and coagulation volume were seen when visible and perfusion-mediated tissue cooling
NaCl was injected (p < .001), with maximum (capillary vascular flow) that functions as a heat
heating and coagulation occurring with 6mL sink. By drawing heat from the treatment zone,
of 36% NaCl solution. Regression analysis this effect reduces the volume of tissue that
demonstrated that both volume and concentra- receives the required minimal thermal dose for
tion of NaCl injected significantly influenced coagulation. Several studies that explored alter-
tissue heating and coagulation. ing tissue perfusion to increase the ablative
In a subsequent study, Lobo et al (76) zone through either mechanical occlusion or
explored the relationship between NaCl pharmacologic agents strongly support the con-
volume and concentration in a reproducible, tention that perfusion-mediated tissue cooling
static agar tissue phantom model. Using a is responsible for this reduction in observed
protein gel with varying NaCl concentration coagulation.
(0-35%) in different-sized wells (0-38cc) sur-
rounding the electrode to simulate tissue injec-
Mechanical Occlusion
tion, various parameters of RF application
(power, current, impedance, temperature) were Using internally cooled electrodes, Goldberg et
recorded. Using mathematical modeling, they al applied RF to normal in vivo porcine liver
derived two equations, one describing the mul- without and with balloon occlusion of the
tivariate relationship to a peak point that rep- portal vein, celiac artery, or hepatic artery, and
resented maximum generator output. The other to ex vivo calf liver (77). Increased coagulation
describes the relationship when temperature was observed with RF ablation combined with
decreases as generator output is exceeded. portal venous occlusion, compared to RF
Both equations predict the effect of volume and without occlusion (P < .01). The differences
concentration of NaCl with an R 2 = .96. Based seen in coagulation correlated to an approxi-
on this, the investigators reexamined the earlier mate reduction in hepatic blood flow.
reported trials in normal porcine liver to corre- Several clinical studies provide additional
late results with these equations. Though the data supporting the role of mechanical blood
constants changed, the equations were able to flow reduction to improve RF ablation efficacy.
account for 86% of the variance seen in those Goldberg et al performed mechanical blood
data. This mathematic characterization is a flow occlusion in three patients with hepatic
significant step toward reliably predicting colorectal metastases undergoing intraopera-
the effects of NaCl when combined with RF tive RF ablation (77). Two similar sized hepatic
ablation. colorectal metastases (2.2-4.2 em) were treated
with identical RF parameters in each patient. In
each case, one lesion was treated with normal
Reducing Blood Flow
blood flow, while the second was treated during
Biophysical aspects of tumor-heat interaction portal flow occlusion (Pringle maneuver) by
must be taken into account when performing clamping the hepatic artery and portal vein to
thermal ablation therapies. Based on the sim- eliminate all intrahepatic blood flow. In each
plified form of the bioheat equation, the third case, coagulation was increased with inflow
component defining thermal induced coagula- occlusion (4.0cm vs 2.5cm, P < .05). Hepatic
tion necrosis is heat loss. Radiofrequency abla- inflow occlusion was performed in additional
tion outcomes in in vivo models have been less patients undergoing intrahepatic RF ablation
successful and more variable compared to for hepatic colorectal metastases. In these cases,
reported reproducible ex vivo results for iden- remote thermometry demonstrated increases in
tical RF protocols. Radiofrequency-induced temperature at 10mm (62°C-72°C) and 20mm
necrosis in vivo is often shaped by the presence (39°C-50°C) distances from the RF probe
3. Image-Guided Tumor Ablation: Basic Science 33

within 5 minutes of portal vein occlusion during Angiographic balloon occlusion can be used,
constant RF application. This increase at the 20 but may not prove adequate for intrahe-
mm distance holds particular significance as the patic ablation given the dual hepatic blood
threshold for induction of coagulation necrosis supply with redirection of compensated flow.
(50°C) was achieved with blood flow reduction. Embolotherapy prior to ablation with particu-
In a second study, Patterson et al have con- lates that occlude sinusoids such as Gelfoam
firmed the strong predictive nature of hepatic and Lipiodol may overcome this limitation, as
blood flow on the extent of RF induced coagu- has been reported by Rossi et al (80). Pharma-
lation in normal in vivo porcine liver using a cologic modulation of blood flow and anti-
hooked electrode system (78). The coagulated angiogenesis therapy are theoretically pos-
focus created by RF during a Pringle maneuver sible, but should currently be considered ex-
was significantly larger in all three dimensions perimental. Another proposal is to combine
than coagulation with unaltered blood flow. RF with arsenic trioxide to increase ablation
Minimum and maximum diameters were sig- efficacy via decreased blood flow.
nificantly increased from 1.2cm to 3.0cm and
3.1 cm to 4.5 cm, respectively, when the Pringle
maneuver was performed (p:::: .002). Coagula-
Microwave
tion volume was increased from 6.5 cm3 to 35.0
Microwaves are a second thermal energy
cm3 with hepatic inflow occlusion. Additionally,
source that has been used for percutaneous
the number of blood vessels within a 1cm
image-guided tumor ablation, and thus far
radius of the electrode strongly predicted
largely used in the Far East. In contrast to RF,
minimum lesion size and lesion volume. In a
in which the inserted electrode functions as the
subsequent study in an in vivo porcine model,
active source, in microwave ablation the
Lu et al examined the effect of hepatic vessel
inserted probes (usually 14 gauge) function as
diameter on RF ablation outcome (79). Using
antennae for externally applied energy at 1000
CT and histopathologic analyses, the authors
to 2450mHz (81). The microwave energy
documented more complete thermal heating
applied to the tissues results in rotation of polar
and a reduced heat-sink effect when hepatic
molecules that is opposed by frictional forces.
vessels within the heating zone were <3mm in
As a result, there is conversion of rotational
diameter. In contrast, vessels >3 mm in diame-
energy into heat (82). One potential advant-
ter had higher patency rates, less endothelial
age of microwave over RF and laser is that
injury, and greater viability of surrounding
investigational ex vivo studies have shown
hepatocytes after RF ablation.
greater tissue penetration and larger zones of
coagulation-necrosis with microwave technol-
Pharmacologic Modulation of Blood Flow ogy. This finding is most pronounced with
cooling of the microwave antenna in a fashion
Goldberg et al modulated hepatic blood flow
similar to cooled-tip RF (83). In clinical prac-
using intraarterial vasopressin and high dose
tice, however, arrays of microwave antennas
halothane in conjunction with RF ablation in in
or multiple insertions of a single microwave
vivo porcine liver (74). Laser Doppler
probe have been necessary to treat lesions
techniques identified a 33 % increase and
greater than 2cm in diameter (84-87). How-
66% decrease in hepatic blood flow after
ever, research likely will adapt advances in RF
administration of vasopresson and halothane,
technology to microwave systems, such as cool-
respectively. Correlation of blood flow to coag-
ing, pulsing, and higher power (83,88,89).
ulation diameter produced was excellent as
R 2 :::: .78.
Clearly, several strategies to reduce blood Laser
flow during ablation therapy have been pro-
posed. Total portal inflow occlusion (Pringle Laser ablation or laser photocoagulation is
maneuver) has been used at open laparotomy. another method to induce thermally mediated
34 M. Ahmed and S.N. Goldberg

coagulation necrosis that has been employed Using a 4-MHz transducer and a power
for percutaneous tumor ablation. For this pro- intensity of 550W/cm2 for 5 seconds, tempera-
cedure, flexible thin optic fibers are inserted ture in excess of 80 a C can be produced in rat
into the target through percutaneously placed liver (101). Areas of coagulation necrosis have
needles using imaging guidance. The laser pro- been shown at histopathology to have a spa-
vides sufficient energy to allow for significant tially sharp demarcation between regions of
heat deposition surrounding the fiber tip, induc- normal and necrotic tissue. By using a suitable
ing protein denaturation and cellular death acoustic frequency, regions of tissue destruction
(90). As with RF systems, thermal profiles have can be induced at depths of up to at least lOcm
been demonstrated to correlate well with the with exposure times on the order of 1 second
extent of coagulation necrosis that is observed (102). One main potential benefit of HIFU is
histopathologically (91,92), as well as with that the focused ablative energy can destroy a
ultrasound (92,93) and Tl-weighted MRI selected target without causing damage to the
(94,95). intervening tissues. Although hampered by
Most devices use a standard laser source small focal zones in the past, this technique is
(neodymium: yttrium-aluminum-garnet [Nd: gaining further enthusiasm as larger zones of
YAG], erbium, holmium, etc.) that produces coagulation are produced by using array trans-
precise wavelengths. Additional device modifi- ducers and other advancing technologies. Fur-
cations on a conventional laser applicator thermore, since insertion of a percutaneous
device have been developed to increase the size probe is not required, HIFU can be considered
of the heated volume. These developments the least invasive of the minimally invasive
include: (a) different types of laser fibers (flex- therapies.
ible/glass dome); (b) modifications to the tip
(i.e., flexible diffusor tip or scattering dome); (c)
length of applicator and diameter of the optic Combination Therapies
fiber; and (d) the number of laser applicators
used (i.e., single versus multiple applicators) The ultimate goal of tumor therapy is complete
(96). Similar to RF technologies, additional eradication of all malignant cells within a focal
device modifications such as pulsing algorithms tumor. Given the high likelihood of incomplete
and internal cooling of the applicator have treatment by heat-based modalities alone, the
been reported (97). case for combining thermal ablation with other
therapies such as chemotherapy or chemoem-
bolization cannot be overstated. The belief that
Ultrasound tumors can reliably destroyed using only one
High-intensity focused ultrasound (HIFU) is a technique is likely overly optimistic, given the
transcutaneous technique that has been studied variety of tumor types and organ sites. Combi-
as a potential method for minimally invasive nation therapy is a key focus of current abla-
treatment of localized benign and malignant tion research. A multidisciplinary approach
tumors (98,99). This technique uses a parabolic that includes surgery, radiation, and chemo-
transducer to focus the ultrasound energy at a therapy is used for the treatment of most solid
distance that creates a focused beam of energy cancers.
with very high peak intensity. This focusing of
energy has been likened to using a magnifying
glass to focus sunlight (100). The focused
Thermal Ablation with Adjuvant
energy is transmitted transcutaneously into the
Chemotherapy
target tissue without requiring percutaneous Strategies that decrease tumor tolerance to
insertion of an electrode or transducer. The heat are not well studied. Theoretically, previ-
ultrasound energy is absorbed by tissue and is ous insult to the tumor cells by cellular hypoxia
converted to heat. Ablation occurs by coagula- (caused by vascular occlusion or antiangiogen-
tion necrosis. esis factor therapy), or prior tumor cell damage
3. Image-Guided Tumor Ablation: Basic Science 35

from chemotherapy or radiation could be used (6.7mm) or RF plus direct intratumoral free
to increase tumor sensitivity to heat (9,10). doxorubicin injection (11.4mm) (p < .01). In
Alternatively, tumor cells that undergo heat- more recent follow-up studies in the same
induced reversible cell injury may demon- animal tumor model, Monsky et al (106) docu-
strate increased susceptibility to secondary mented a fivefold increase in intratumoral
chemotherapy. Synergy between chemotherapy doxorubicin concentration for tumors receiving
and hyperthermic temperatures (42-45°C) has RF ablation, while D'Ippolito et al (107)
been established (44,103). demonstrated reduced tumor growth in animals
In one study, Goldberg et al (104) treated 1.2- receiving combination RF/Doxil treatment
to 1.5 cm R3230 rat mammary adenocarcinoma over either RF ablation or Doxil alone.
with RF and/or intratumoral injection of dox- Recently, Goldberg et al (108) conducted a
orubicin chemotherapy. Tumors were treated pilot clinical study combining RF ablation with
with combinations of RF alone (monopolar, adjuvant Doxil therapy in 10 patients with liver
70°C for 5 minutes), direct intratumoral dox- malignancies. Patients with at least one liver
orubicin injection (250 J..lL; 0.5 mg in total) lesion (mean size 4.0 ± 1.8cm) were random-
alone, intratumoral doxorubicin followed by ized into two groups for treatment with RF
RF, and no treatment. RF alone, RF with alone (n = 5) versus RF with pretreatment
distilled water, and intratumoral doxoru- single-dose intravenous Doxil (24 hours pre-
bicin alone produced 6.7mm, 6.9mm, and 2mm RF, 20mg/kg). Several tumor types were
of coagulation, respectively. Significantly treated, including primary hepatocellular carci-
increased coagulation occurred with combined noma (n =4), colorectal metastases (n =3), neu-
RF and intratumoral doxorubicin (11.4mm; roendocrine tumors (n = 2), and breast cancer
p < .001). Additional experiments conducted to metastases (n = 1). Patients receiving Doxil
examine the effect and timing of doxorubicin therapy with RF ablation demonstrated a 25%
adminstration demonstrated greatest effect increase in coagulation volume 2 to 4 weeks
when the drug was given 30 minutes after RF postablation. Two Doxil/RF ablation-treated
application. In particular, the increased necro- tumors with incomplete initial treatment
sis seen when the doxorubicin was injected denoted by contrast enhancement at the
after RF points to a potential two-hit effect, periphery of the ablation zone on baseline post-
with initial reversible cell injury inflicted by RF scans were converted to complete ablation
sublethal doses of heat in the more peripheral within 4 weeks of treatment. Similar incom-
ablation zone, followed by irreversible injury by pletely ablated tumors treated with RF alone
doxorubicin on already susceptible cells. did not show a similar conversion. Additionally,
Subsequently, investigators explored intra- in the tumors treated incompletely with combi-
venous doxorubicin use as a means to achieve nation Doxil and RF ablation, large blood
greater delivery to the zone surrounding RF- vessels traversing the ablation zone were
induced coagulation. Recent study has focused present on baseline scans, but were absent on
on the use of a commercially available, steri- follow-up imaging. These preliminary results
cally stabilized liposomal preparation of highlight the potential of combining RF
doxorubicin (Doxil, Alza Pharmaceuticals). ablation with adjuvant liposomal chemothera-
Liposomes function as delivery vehicles for a peutic agents for greater and more complete
variety of chemotherapeutic agents through treatment.
increased circulating time and greater tumor Several separate mechanisms likely underlie
specificity, with the added advantage of reduced this considerable synergy between RF and
drug toxicity. Goldberg et al (105) combined Doxil. Postulated reasons for this adjuvant
RF ablation with intravenous Doxil (0.5mL, effect can be divided into two distinct cate-
1.0mg in total) in a rat mammary adenocarci- gories. The first involves increased delivery of
noma model. A significant increase in coagula- Doxil into tumor tissue peripheral to the
tion (13.1 mm) was seen with this particular central RF coagulation zone. Monsky et al
approach compared to either RF alone (106) explored intratumoral distribution using
36 M. Ahmed and S.N. Goldberg

radiolabeled liposomal doxorubicin prepara- ize many of the basic principles underlying the
tions identical to Doxil. They were able to visu- tumor ablative features of these treatments. We
alize the distribution of liposomes within the have provided an overview of the basic princi-
tumor post-RF ablation, which concentrated in ples of each of the widely used strategies and
a thick rim of tumor surrounding the central RF described the recent technologic modifications
zone. Quantitation of doxorubicin content of that have been developed to further improve
RF-treated tumors compared to untreated clinical success of these therapies. Future direc-
tumors revealed a fivefold increase in Doxil tions of research are now looking to combina-
uptake (p < .01). Potential explanations for this tion therapies, such as combining liposomal
increased delivery include nonspecific RF- chemotherapy with thermal ablation, as the
induced inflammation in surrounding tissue next step to further improve the clinical effec-
leading to hyperthermic vasodilation and tiveness of such therapies.
increased vascular permeability and increased
cellular uptake and retention of doxorubicin
due to disruption of cellular defense mecha-
References
nisms. Kruskal et al (109) used dynamic intra-
vital video microscopy to study microvascular 1. Dodd GD III, Soulen MC, Kane RA, et aL Min-
and cellular alterations in normal livers of imally invasive treatment of malignant hepatic
live nude mice treated with RF ablation. At tumors: at the threshold of a major break-
temperatures well below those required for through. Radiographies 2000;20:9-27.
irreversible thermal damage, several distinct 2. Goldberg SN, Gazelle GS, Mueller PRo Thermal
ablation therapy for focal malignancy: a unified
changes conducive to increased liposome
approach to underlying principles, techniques,
delivery were identified beyond the central and diagnostic imaging guidance. Am J Radiol
coagulation necrosis, including alterations in 2000;174:323-331.
permeability, reversible microvascular stasis, 3. Goldberg SN, Dupuy DE. Image-guided
and increased endothelial leakiness. radiofrequency tumor ablation: challenges and
A second likely means of synergy between opportunities-Part I. J Vase Intervent Radiol
RF ablation and Doxil concerns the tumorici- 2001;12:1021-1032.
dal effect of both the liposome and doxorubicin 4. Colella G, Bottelli R, De Carlis L, et al. Hepa-
components of Doxil. Interestingly, tumors tocellular carcinoma: comparison between
treated with RF in combination with empty liver transplantation, resective surgery, ethanol
liposome preparations (identical formulation injection, and chemoembolization. Transpl Int
1998;11(suppll):SI93-196.
to Doxil without doxorubicin) also demon-
5. Liver Cancer Study Group of Japan. Survey and
strated significantly greater necrosis than RF follow-up study of primary liver cancer in
alone (lO.9mm, p < .01) (105). Prior studies Japan: report 14. Kyoto: Shinko-Insatsu, 2000.
investigating synergy between lipid prepara- 6. Ahmed M, Goldberg SN. Thermal abla-
tions and low-level hyperthermia have identi- tion therapy for hepatocellular carcinoma.
fied free radical generation as a cause of J Vase Intervent Radiol2oo2;13(9 suppl):S231-
cellular injury (110). Further research may 244.
focus on optimizing liposome structure to 7. Pautler SE, Pavlovich CP, Mikityansky I, et al.
foster this effect for greater RF coagulation Retroperitoneoscopic-guided radiofrequency
gain. ablation of renal tumors. Can J Urol 2001;8:
1330-1333.
8. Pavlovich CP, Walther MM, Choyke PL, et aL
Percutaneous radio frequency ablation of small
Conclusion renal tumors: initial results. J Urol 2002;167:
10-15.
Several different percutaneous and minimally 9. Dupuy DE, Zagoria RJ, Akerley W, Mayo-
invasive therapies, both chemical and thermal, Smith WW, Kavanaugh PV, Safran H. Percuta-
have been well described to treat focal malig- neous RF ablation of malignancies in the lung.
nancies. Investigators have begun to character- AJR 2000;174:57--60.
3. Image-Guided Tumor Ablation: Basic Science 37

10. Zagoria RJ, Chen MY, Kavanagh PV, Torti FM. guided intratumoral injection of acetic acid.
Radio frequency ablation of lung metastases Radiology 1994;193:747-752.
from renal cell carcinoma. J Urol 2001;166: 23. Ohnishi K, Yoshioka H, Ito S, Fujiwara K.
1827-1828. Prospective randomized controlled trial com-
11. Woertler K, Vestring T, Boettner F, Winkel- paring percutaneous acetic acid injection and
mann W, Heindel W, Lindner N. Osteoid percutaneous ethanol injection for small hepa-
osteoma: CT-guided percutaneous radiofre- tocellular carcinoma. Hepatology 1998;27:
quency ablation and follow-up in 47 patients. 67-72.
J Vasc Intervent RadioI2001;12:717-722. 24. Cooper IS. Cryogenic surgery: a new method of
12. Jeffery SS, Birdwell RL, Ikeda DM. Radio- destruction or extirpation of benign or malig-
frequency ablation of breast cancer: first report nant tissue. N Engl J Med 1963;268:743-749.
of an emerging technology. Arch Surg 1999;134: 25. Onik G, Kane RA, Steele G. Monitoring
1064-1068. hepatic cryosurgery with sonography. AJR
13. Livraghi T, Giorgio A, Marin A. Hepatocellular 1986;147:665-669.
carcinoma and cirrhosis in 746 patients: long- 26. Lee FT, Mahvi DM, Chosy SG, et a1. Hepatic
term results of percutaneous ethanol injection. cryosurgery with intraoperative US guidance.
Radiology 1995;197:101-108. Radiology 1997;202:624-632.
14. Shiina S, Tagawa K, Niwa Y. Percutaneous 27. Cozzi PJ, Stewart GJ, Morris DL. Thrombocy-
ethanol injection therapy for hepatocellular topenia after hepatic cryosurgery for colorectal
carcinoma: results in 146 patients. AJR 1993; metastases: correlates with hepatic injury.
160:1023-1025. World J Surg 1994;18:774-777.
15. Kawano M. An experimental study of percuta- 28. Rubinsky B, Lee CY, Bastacky J, Onik G. The
neous absolute ethanol injection therapy for process of freezing and the mechanism of
small hepatocellular carcinoma: effects of damage during hepatic cryosurgery. Cryobiol-
absolute ethanol on healthy canine liver. ogy 1990;27:85-97.
Gastroenterol Jpn 1989;24:663-669. 29. Silverman SG, Thncali K, Adams DF, et a1. MR
16. Shiina S, Tagawa K, Unuma T. Percutaneous image-guided percutaneous cryotherapy of
ethanol injection therapy for hepatocellular liver tumors: initial experience. Radiology 2000;
carcinoma: a histopathologic study. Cancer 217:657-664.
1991;68:1524-1530. 30. Hoffmann NE, Bischof Je. The cryobiology of
17. Giovannini M, Seltx UF. Ultrasound guided cryosurgical injury. Urology 2002;60:40-49.
percutaneous alcohol injection of small liver 31. Mazur P. Freezing of living cells: mechanisms
metastases. Cancer 1994;73:294-297. and implications. Am J Physiol 1984;247:125-
18. Livraghi T, Vettori C, Torzilli G, Lazzaroni S, 142.
Pellicano S, Ravasi S. Percutaneous ethanol 32. Lovelock JE. The hemolysis of human red
injection of hepatic tumors: single-session blood cells by freezing and thawing. Biochem
therapy under general anesthesia. AJR 1993; Biophys Acta 1953;10:414-426.
160:1065-1069. 33. Steponkus PL. Role of plasma membrane in
19. Livraghi T. Percutaneous ethanol injection in freezing injury and cold acclimation. Annu Rev
the treatment of hepatocellular carcinoma in Plant PhysioI1984;35:543-584.
cirrhosis. Hepatogastroenterology 2001;48: 34. Toner M. Nucleation of ice crystals inside bio-
20-24. logical cells. In: Steponkus PL, ed. Advances in
20. Liang HL, Yang CF, Pan HB, et a1. Small Low-Temperature Biology. 1993:1-52.
hepatocellular carcinoma: safety and efficacy 35. Pollock GA, Pegg DE, Hardie IR. An isolated
of single high-dose percutaneous acetic acid perfused rat mesentery model for direct obser-
injection for treatment. Radiology 2000;214: vation of the vasculature during cryopreserva-
769-774. tion. Cryobiology 1986;23:500-511.
21. Ohnishi K, Yoshioka H, Ito S, Fujiwara K. 36. Hoffmann NE, Bischof Je. Cryosurgery of
Treatment of nodular hepatocellular carcinoma normal and tumor tissue in the dorsal skin flap
larger than 3 cm with ultrasound-guided percu- chamber. II. J Biomech Eng 2001;123:310-316.
taneous acetic acid injection. Hepatology 37. Barker JH, Bartlett R, Funk W. The effect of
1996;24:1379-1385. superoxide dismutase on ths skin microcircula-
22. Ohnishi K, Ohyama N, Ito S, Fujiwara K. Small tion after ischemia and reperfusion. Prog Appl
hepatocellular carcinoma: treatment with US- Microcirc 1987;12:276-281.
38 M. Ahmed and S.N. Goldberg

38. Zook N, Hussmann J, Brown R. Microcircula- treatment of hepatic cancer. AJR 1996;167:
tory studies of frostbite injury. Ann Plast Surg 759-768.
1998;40:246-253. 52. Solbiati L, Ierace T, Goldberg SN. Percutaneous
39. Asahina E, Shimada K, Hisada Y. A stable state US-guided RF tissue ablation of liver metas-
of frozen protoplasm with invisible intracellu- tases: long-term follow-up. Radiology 1997;202:
lar ice crystals obtained by rapid cooling. Exp 195-203.
Cell Res 1970;59:349-358. 53. Goldberg SN, Gazelle GS, Dawson SL, Rittman
40. Gazelle GS, Goldberg SN, Solbiati L, Livraghi WJ, Mueller PR, Rosenthal DI. Tissue ablation
T. Tumor ablation with radiofrequency energy. with radiofrequency using multiprobe arrays.
Radiology 2000;217:6333-6346. Acad RadioI1995;2:670-674.
41. McGahan Jp' Dodd GD III. Radiofrequency 54. Rossi S, Buscarini E, Garbagnati F. Percuta-
ablation of the liver: current status. AJR 2001; neous treatment of small hepatic tumors by an
176:3-16. expandable RF needle electrode. AJR 1998;
42. Cosman E, Nashold B, Ovelman-Levitt J. The- 170:1015-1022.
oretical aspects of radiofrequency lesions in the 55. Siperstein AE, Rogers SJ, Hansen PD,
dorsal root entry zone. Neurosurgery 1984;15: Gitomirsky A. Laparoscopic thermal ablation
945-950. of hepatic neuroendocrine tumor metastases.
43. Seegenschmiedt M, Brady L, Sauer R. Intersti- Surgery 1997;122:1147-1155.
tial thermoradiotherapy: review on technical 56. Leveen RF. Laser hyperthermia and radio-
and clinical aspects. Am J Clin Oncol 1990;13: frequency ablation of hepatic lesions. Semin
352-363. Intervent RadioI1997;12:313-324.
44. Trembley B, Ryan T, Strohbehn J. Interstitial 57. Berber E, Foroutani A, Garland AM, et al.
hyperthermia: physics, biology, and clinical Use of CT Hounsfield unit density to iden-
aspects. In: Hyperthermia and Oncology, vol 3. tify ablated tumor after laparoscopic radiofre-
Utrecht: YSP, 1992:11-98. quency ablation of hepatic tumors. Surg Endosc
45. Larson T, Bostwick D, Corcia A. Temperature- 2000;14:799-804.
correlated histopathologic changes following 58. de Baere T, Denys A, Johns Wood B, et al.
microwave thermoablation of obstructive Radiofrequency liver ablation: experimental
tissues in patients with benign prostatic comparative study of water-cooled versus
hyperplasia. Urology 1996;47:463--469. expandable systems. AJR 2001;176:187-192.
46. Zervas N, Kuwayama A. Pathologic analysis of 59. McGahan JP, Gu WZ, Brock JM, Tesluk H,
experimental thermal lesions: comparison of Jones CD. Hepatic ablation using bipolar
induction heating and radiofrequency electro- radiofrequency electrocautery. Acad Radiol
coagulation. J Neurosurg 1972;37:418--422. 1996;3:418--422.
47. Thomsen S. Pathologic analysis of photother- 60. Desinger K, Stein T, Muller G, Mack M, YogI
mal and photomechanical effects of laser tissue T. Interstitial bipolar RF-thermotherapy
interactions. Photochem Photobiol 1991;53: (REITT) therapy by planning by computer
825-835. simulation and MRI-monitoring-a new
48. Goldberg SN, Gazelle GS, Compton CC, concept for minimally invasive procedures.
Mueller PRo Tanabe KK. Treatment of intra- Proc SPIE 1999;3249:147-160.
hepatic malignancy with radiofrequency abla- 61. Haemmerich DG, Lee FrJ, Chachati L, Wright
tion: radiologic-pathologic correlation. Cancer AS, Mahvi DM, Webster JG. A device that
2000;88:2452-2463. allows for multiple simultaneous radiofre-
49. Goldberg SN, Gazelle GS, Halpern EF, Rittman quency (RF) ablations in separated areas of the
WJ, Mueller PR, Rosenthal DI. Radiofre- liver with impedance-controlled cool-ip probes:
quency tissue ablation: importance of local tem- an ex vivo feasibility study [abstract]. Radiol-
perature along the electrode tip exposure in ogy 2002;225(p):242.
determining lesion shape and size. Acad Radiol 62. Haemmerich DG, Lee FrJ, Mahvi DM, Wright
1996;3:212-218. AS, Webster Jo. Multiple probe radiofre-
50. Pennes H. Analysis of tissue and arterial blood quency: rapid switching versus simultaneous
temperatures in the resting human forearm. power application in a computer model. Radi-
J Appl PhysioI1948;1:93-122. ology 2002;225(p):639.
51. Rossi S, DiStasi M, Buscarini E. Percuta- 63. Goldberg SN, Gazelle GS, Solbiati L, Rittman
neous RF interstitial thermal ablation in the WJ, Mueller PRo Radiofrequency tissue abla-
3. Image-Guided Tumor Ablation: Basic Science 39

tion: increased lesion diameter with a perfusion 75. Merkle E, Goldberg SN, Boll DT, et a1. Effect
electrode. Acad Radiol 1996;3:636-644. of supramagnetic MR contrast agents on radio-
64. Lorentzen T. A cooled needle electrode for frequency induced temperature distribution: in
radiofrequency tissue ablation: thermodynamic vitro measurements in polyacrylamide phan-
aspects of improved performance compared toms and in vivo results in a rabbit liver model.
with conventional needle design. Acad Radiol Radiology 1999;212:459--466.
1996;3:556-563. 76. Lobo SM, Afzal SK, Kruskal JB, Lenkinski RE,
65. Solbiati L, Ierace T, Tonolini M, Osti V, Cova L. Gazelle GS, Goldberg SN. Radiofrequency
Radiofrequency thermal ablation of hepatic thermal ablation using an adjuvant NaCl gel:
metastases. Eur J Ultrasound 2001;13:149-158. effect of electrical conductivity on tissue coag-
66. Goldberg SN, Solbiati L, Hahn PF, et a1. Large- ulation [abstract]. Radiology 2001;201 (suppl):
volume tissue ablation with radiofrequency by 398.
using a clustered, internally-cooled electrode 77. Goldberg SN, Hahn PF, Tanabe KK, et al.
technique: laboratory and clinical experience Percutaneous radiofrequency tissue ablation:
in liver metastases. Radiology 1998;209:371- does perfusion-mediated tissue cooling limit
379. coagulation necrosis? J Vasc Intervent Radiol
67. Goldberg SN, Stein M, Gazelle GS, Sheiman 1998;9:101-111.
RG, Kruskal JB, Clouse ME. Percutaneous 78. Patterson EJ, Scudamore CH, Owen DA, Nagy
radiofrequency tissue ablation: optimization of AG, Buczkowski AK. Radiofrequency ablation
pulsed-RF technique to increase coagulation of porcine liver in vivo: effects of blood flow
necrosis. J Vase Intervent Radiol 1999;10:907- and treatment time on lesion size. Ann Surg
916. 1998;227:559-565.
68. Curley MG, Hamilton PS. Creation of large 79. Lu DS, Raman SS, Vodopich DJ, Wang M, Sayre
thermal lesions in liver using saline-enhanced J, Lassman C. Effect of vessel size on creation
RF ablation. Proc 19th International Confer- of hepatic radiofrequency lesions in pigs:
ence IEEE/EMBS 1997:2516-2519. assessment of the "heat sink" effect. AJR 2002;
69. Livraghi T, Goldberg SN, Monti F, et a1. Saline- 178:47-51.
enhanced radiofrequency tissue ablation in the 80. Rossi S, Garbagnati F, Lencioni R, et a1. Percu-
treatment of liver metastases. Radiology 1997; taneous radiofrequency thermal ablation of
202:205-210. nonresectable hepatocellular carcinoma after
70. Miao Y, Ni Y, Yu J, Marchal G. A comparative occlusion of tumor blood supply. Radiology
study on validation of a novel cooled-wet elec- 2000;217:119-126.
trode for radiofrequency liver ablation. Invest 81. King RWP, Shen LC, Wu IT. Embedded insu-
Radiol 2000;35:438--444. lated antenna for communication and heating.
71. Miao Y, Ni Y, Yu J, Zhang H, Baert A, Marchal Electromagnetics 1981;1:51-72.
G. An ex vivo study on radiofrequency tissue 82. Foster KR, Schepps JL. Dielectric properties of
ablation: increased lesion size by using an tumor and normal tissues at radio through
"expandable-wet" electrode. Eur Radiol 2001; microwave frequencies. J Microwave Power
11:1841-1847. 1981;16:107-119.
72. Kettenbach J, Kostler W, Rucklinger E, et a1. 83. Moriyama E, Matsumi N, Shiraishi T, et a1.
Percutaneous saline-enhanced radiofrequency Hyperthermia for brain tumors: improved
ablation of unresectable liver tumors: initial delivery with a new cooling system. Neuro-
experience in 26 patients. AJR 2003;180:1537- surgery 1988;23:189-195.
1545. 84. Saitsu H, Mada Y, Taniwaki S, et a1. Investiga-
73. Goldberg SN, Ahmed M, Gazelle GS, et a1. tion of microwave coagulo-necrotic therapy for
Radiofrequency thermal ablation with adju- 21 patients with small hepatocellular carcinoma
vant saline injection: effect of electrical con- less than 5 cm in diameter. Nippon Geka
ductivity on tissue heating and coagulation. Gakkai Zasshi 1993;94:356-365.
Radiology 2001;219:157-165. 85. Saitsu H, Nakayama T. Microwave coagulo-
74. Goldberg SN, Hahn PF, Halpern EF, Fogle R, necrotic therapy for hepatocellular carcinoma.
Gazelle GS. Radiofrequency tissue ablation: Nippon Rinsho 1993;51:1102-1107.
effect of pharmacologic modulation of blood 86. Watanabe Y, Sato M, Abe Y, et a1. Laparoscopic
flow on coagulation diameter. Radiology 1998; microwave coagulo-necrotic therapy for hepa-
209:761-769. tocellular carcinoma: a feasible study of an
40 M. Ahmed and S.N. Goldberg

alternative option for poor-risk patients. corporeal high-intensity focused ultrasound.


J Laparoendosc Surg 1995;5:169-175. Eur Urol 1993;23(suppl 1):17-22.
87. Sato M, Watanabe Y, Ueda S, et al. Microwave 99. Jolesz FA, Hynynen K. Magnetic resonance
coagulation therapy for hepatocellular carci- image-guided focused ultrasound surgery.
noma. Gastroenterology 1996;110:1507-1514. Cancer 2002;8(suppll):SlOQ-112.
88. Shibata T, Emuro Y, Yamamoto Y, et al. Small 100. Sanghvi NT, Hawes RH. High-intensity focused
hepatocellular carcinoma: comparison of radio- ultrasound. Gastrointest Endosc Clin North
frequency ablation and percutaneous micro- Am 1994;4:383-395.
wave coagulation therapy. Radiology 2002;223: 101. Reilly CR, Yang R, Reilly WM. Tissue heating
331-337. measurements during high intensity focused
89. Dong B, Liang P, Yu X, et al. Percutaneous ultrasound cancer therapy. J Ultrasound Med
sonographically guided microwave coagulation 1991;1O:S26.
therapy for hepatocellular carcinoma: results in 102. Hill CR, ter Haar GR. Review article: high
234 patients. AJR 2003;180:1547-1555. intensity focused ultrasound-potential for
90. Brown S. Laser-tissue interactions. Krausner N cancer treatment. Br J Radiol 1995;68:1296-
1991;Lasers in Gastroenterology:37-50. 1303.
91. Dachman AH, McGehee JA, Beam TE, Burris 103. Christophi C, Muralidharan V. Treatment of
JA, Powell DA. US-guided percutaneous laser hepatocellular carcinoma by percutaneous
ablation of liver tissue in a chronic pig model. laser hyperthermia. J Gastroenterol Hepatol
Radiology 1990;176:129-133. 2001;16:548-552.
92. Jiao LR, Hansen PD, Havlik R, Mitry RR, 104. Goldberg SN, Saldinger PF, Gazelle GS, et al.
Pignatelli M, Habib N. Clinical short-term Percutaneous tumor ablation: increased coagu-
results of radiofrequency ablation in primary lation necrosis with combined radiofrequency
and secondary liver tumors. Am J Surg 1999; and percutaneous doxorubicin injection. Radi-
177:303-306. ology 2001;220:420-427.
93. Nolsoe CP, Torp-Pedersen S, Burcharth F, et al. 105. Goldberg SN, Girnan GD, Lukyanov AN, et al.
Interstitial hyperthermia of colorectal liver Percutaneous tumor ablation: increased necro-
metastases with an US-guided Nd-YAG laser sis with combined radio-frequency ablation and
with a diffuser tip: a pilot clinical study. Radi- intravenous liposomal doxorubicin in a rat
ology 1993;187:333-337. breast tumor model. Radiology 2002;222:797-
94. Diederich CJ, Nau WH, Deardorff DL. Pros- 804.
tate thermal therapy with interstitial and 106. Monsky WL, Kruskal JB, Lukyanov AN, et al.
transurethral ultrasound applicators: a feasibil- Radio-frequency ablation increases intratu-
ity study. In: Ryan TP, ed. Surgical Applications moralliposomal doxorubicin accumulation in a
of Energy, Proceedings of SPIE Vol. 3249. rat breast tumor model. Radiology 2002;224:
1998:2-13. 823-829.
95. Vogi TJ, Muller PK, Hammerstingl R, et al. 107. D'Ippolito G,Ahmed M, Girnan GD, et al. Per-
Malignant liver tumors treated with MR cutaneous tumor ablation: increased endpoint
imaging-guided laser-induced thermotherapy: survival with combined radiofrequency abla-
technique and prospective results. Radiology tion and liposomal doxorubicin in a rat breast
1995;196:257-265. tumor model. Radiology; in press.
96. Vogi T, Eichler K, Straub R, et al. Laser-induced 108. Goldberg SN, Kamel IR, Kruskal JB, et al.
thermotherapy of malignant liver tumors: gen- Radiofrequency ablation of hepatic tumors:
eral principles, equipment, procedure-side increased tumor destruction with adjuvant
effects, complications and results. Eur J Ultra- liposomal doxorubicin therapy. AJR 2002;179:
sound 2001;13:117-127. 93-101.
97. Vogi TJ, Mack MG, Roggan A, et al. Internally 109. Kruskal JB, Oliver B, Huertas JC, Goldberg SN.
cooled power laser for MR-guided interstitial Dynamic intrahepatic flow and cellular alter-
laser-induced thermotherapy of liver lesions: ations during radiofrequency ablation of liver
initial clinical results. Radiology 1998;209: tumors in mice. J Vasc Intervent Radiol 2001;
381-385. 12:1193-1201.
98. Yang R, Sanghvi NT, Rescorla FJ, Kopecky KK, 110. Kong G, Dewhirst MW. Hyperthermia and lipo-
Grosfeld JL. Liver cancer ablation with extra- somes. Int J Hyperthermia 1999;15:345-370.
4
Tumor Angiogenesis: General
Principles and Therapeutic Approaches
John V. Heymach and Judah Folkman

Tumor growth is dependent on angiogenesis, a transparent rabbit ear chamber, Ide and
the process by which new (;apillary blood colleagues (4) showed in 1939 that wound-
vessels are recruited and sustained. In recent associated vessels regressed during the healing
years, key steps in the angiogenic process process, whereas a tumor implant caused pro-
have been identified, and various angiogenesis gressive growth of new vessels. Algire and
inhibitors have been developed. These agents coworkers (5) showed in the mid-1940s that
are now in clinical testing for cancer and a new vessels in the neighborhood of a tumor
number of other diseases. In the majority of implant arose from existing vessels in the host,
cases, angiogenesis inhibitors are being tested and not from the tumor itself. These studies
alone and in combination with chemotherapy were largely overlooked, and it was generally
for advanced or metastatic cancers. Recent assumed that tumors could expand by simply
evidence suggests, however, that they poten- living on preexisting vessels. Tumor-induced
tially may be beneficial in earlier stage disease changes such as those seen in the rabbit ear
(i.e., chemoprevention) or in combination with chamber were assumed to be a nonspecific
other modalities such as radiation therapy. Fur- inflammatory reaction to the tumor (6).
thermore, tumor ablation techniques such as In the 1960s, it was shown that tumor growth
hyperthermia and chemoembolization work, at in an isolated organ perfusion system was
least in part, through effects on the tumor vas- severely restricted in the absence of tumor
culature. Understanding mechanisms of tumor neovascularization (7,8). Based on these obser-
angiogenesis, therefore, may shed light on ways vations, in 1971 it was proposed that tumor
to inhibit tumor growth and increase the effec- growth is angiogenesis dependent, and that
tiveness of existing treatment modalities. inhibiting angiogenesis might therefore be a
therapeutic strategy for fighting cancer (9). Fur-
thermore, it was proposed that tumors secreted
Historical Background diffusible factors that activated the normally
quiescent host endothelial cells, stimulating
It was observed more than a century ago that their proliferation and migration. This hypoth-
tumors have a rich vascular supply, and are esis was supported by the observations that
often more vascularized than normal tissues in the absence of neovascularization, tumors
(1,2). This was initially attributed to a simple failed to grow beyond several millimeters and
dilation of existing host blood vessels (3), stim- remained in a dormant state (10,11).
ulated by factors secreted by the tumor. Three Experimental confirmation and acceptance
studies in the 1930s and 1940s suggested that of this hypothesis were slow, as they required a
tumor hyperemia could be related to new blood number of scientific developments, including:
vessel growth stimulated by the tumor. Using (a) the development of in vitro methods for cul-

41
42 IV. Heymach and I Folkman

turing endothelial cells, (b) the discovery of Key Steps in the Angiogenic
angiogenesis inhibitors, and (c) identification of
tumor-derived proteins that stimulated angio- Process
genesis (12-17), particularly basic fibroblast
growth factor (bFGF) and vascular endothelial In the adult, angiogenesis normally occurs
growth factor (VEGF) (16-20). Because of during wound healing and in the female repro-
these early efforts, the nascent field of angio- ductive system. With these exceptions, endothe-
genesis research began and has subsequently lium is typically quiescent with turnover times
undergone explosive growth through the 1980s measured in the hundreds of days. Bone
and 1990s. The field encompasses a broad range marrow cells, in contrast, maintain an average
of basic science and clinical disciplines, with turnover time of less than 1 week. Pathologic
active areas of investigation including oncology, neovascularization occurs in a number of dis-
cardiology, dermatology, gynecology, ophthal- eases, including diabetic retinopathy, rheuma-
mology, and developmental biology. To illus- toid arthritis (21), and cancer.
trate this growth, in 1980 there were fewer than Tumor angiogenesis is a complex, multistep
40 publications with angiogenesis in the title or process regulated by the local balance of
abstract, whereas in 2002 there were approxi- endogenous pro- and antiangiogenic factors
mately 3800. More importantly, there are (22-25). The molecular mechanisms underlying
currently more than 100 clinical trials of these processes are now understood in some
angiogenesis inhibitors for cancer patients in detail, and each step is being investigated as
the United States alone. a potential therapeutic target (Fig. 4.1). The

Tumor

Support cells MMPs Ang-2

8M ~rl~·~~~·~~-I. ;!.!::!Z:t~~::~~·~~!=iJ

- .-
Endothelium

=
';;;,dGJG*Q\ s

~ Normal vessel ... I!] Destabilized ... [£] Endothelial proliferation.... [[) CEP
vessel migration. & assembly; mobilization
angiogenic sprouting from bone marrow

FIGURE 4.1. Steps in tumor angiogenesis. A: Normal (VEGF), endothelial cells proliferate, migrate,
vessel with intact basement membrane, surrounded and assemble into capillary tubes that form tumor
by support cells (i.e., pericyctes, smooth muscle vasculature. D: Bone marrow-derived circulating
cells). B: The tumor stimulates breakdown of the endothelial precursors (CEPs) are mobilized in
basement membrane by matrix metalloproteinases response to VEGF and other factors, and may
(MMPs) and other proteases. Interactions between provide a second source of endothelial cells for
support cells and endothelium are disrupted by tumor vasculature. (Reprinted with permission from
angiopoietin-2 (Ang-2). C: In response to tumor- IV. Heymach. "Angiogenesis and antiangiogenic
derived factors such as basic fibroblast growth factor approaches to sarcomas." Current Opinion in Oncol-
(bFGF) and vasculature endothelial growth factor ogy 2001;13:261-269.)
4. Tumor Angiogenesis 43

TABLE 4.1. Endogenous stimulators and inhibitors of angiogenesis.


Stimulators Inhibitors
Vascular endothelial growth factor-A, E, C Thrombospondin-l,2
Basic fibroblast growth factor Endostatin
Acidic fibroblast growth factor Angiostatin
Transforming growth factor-a. Thmstatin
Transforming growth factor-~ Canstatin
Platelet-derived growth factor Antiangiogenic antithrombin III
Interleukin-8 Interferon-a., ~, y
Hepatocyte growth factor Platelet factor-4
EG-VEGF
Angiopoietin-l
Matrix metalloproteinases (MMPs)
Proangiogenic oncogenes: K-ras, H-ras,
Src, p53, EGFR, Her-2, c-rnyb, etc.

EG-VEGF, endocrine gland-derived vascular endothelial growth factor; EGFR,


epidermal growth factor receptor.

initial step involves degradation of the under- Contribution of Circulating


lying extracellular matrix and basement mem-
brane by matrix metalloproteinases (MMPs), Endothelial Cells to Tumor
particularly MMP-2 and MMP-9, and other Vascularization
proteinases (26,27). The host vessel is further
destabilized by angiopoietin-2 (Ang-2), a In the above model, endothelial cells in tumor
natural antagonist for the endothelial-specific vasculature arise through the proliferation of
Tie-2 receptor (28,29). Tie-2 plays a critical role ECs in local vessels. In the 1990s, however,
in promoting and maintaining the interaction studies by Isner and colleagues (33-35)
between endothelial and supporting cells (30). revealed another potential supply of cells to
Freed from the constraints of their surround- participate in tumor neovascularization: bone
ing basement membrane and support cells, marrow-derived circulating endothelial pre-
endothelial cells then proliferate, migrate, and cursors (CEPs). Using animal models in which
assemble into tubes. A number of molecules bone marrow cells were labeled with lacZ as a
contribute to these processes (Table 4.1), al- marker, CEPs were found to contribute to
though VEGF and bFGF are thought to playa neovascularization occurring during wound
preeminent role. VEGF also acts as a survival healing, limb ischemia, and tumor growth (35).
factor for endothelial cells (Ecs) (31), regulates These cells were mobilized by VEGF (35). The
vascular permeability (32), and promotes the contribution of CEPs to tumor neovasculariza-
recruitment of circulating endothelial precur- tion and growth was further established by a
sors (see below). Migration and assembly are study in which mice rendered incapable of
dependent on adhesion molecules such as undergoing normal angiogenesis through tar-
integrin av~3, vascular endothelial cadherin, geted disruption of one allele of Idl and two
vascular cell adhesion molecule-l (VCAM-l), alleles of Id3 (36). In these mice, three differ-
and members of the selectin family. Finally, ent types of implanted tumors failed to induce
stabilization and maturation of the new vessels angiogenesis, their growth was severely
require recruitment of pericytes and smooth restricted, and they did not metastasize.
muscle cells and production of a new basement However, when these mice were injected with
membrane, processes involving Ang-l, platelet- bone marrow from wild-type mice, circulating
derived growth factor (PDGF), transforming endothelial precursors from the donor marrow
growth factor-~ (TGF-~), and other factors were recruited to the tumor vascular bed and
(23,25,29,30). restored the ability of these tumors to undergo
44 IV. Heymach and I Folkman

angiogenesis and grow at essentially the same tribution is likely to depend on the specific
rate as in wild-type mice (37). This established tumor type and local balance of angiogenic
that bone marrow--derived cells were capable factors.
of differentiating into endothelial cells, becom-
ing tumor vasculature, and restoring tumor
growth. Subsequent animal studies suggest that Role of Angiogenesis in
tumor types vary in their ability to recruit and
incorporate circulating endothelial precursors Tumor Progression and
(38). Metastatic Spread
In humans, the contribution of bone
marrow-derived endothelial cells to tumor vas- The development of cancer is a multistage
culature in humans remains to be determined. process, and angiogenesis may play an impor-
Circulating endothelial cells can be detected tant role at one or more points in the devel-
in the blood using cell surface markers (39), opment of cancer (Fig. 4.2). During the
and they are known to be elevated in cancer premalignant (avascular) phase, when angio-
patients (40,41) and to change in response to genic activity is absent or insufficient, tumors
antiangiogenic therapy (41), raising the possi- remain small (1-2mm 3). These premalignant
bility that they may be useful as a surrogate lesions, which may be categorized as hyper-
marker for trials of angiogenesis inhibitors plasia, metaplasia, dysplasia, or carcinoma in
(42). They are also elevated in patients with situ, often have genetic changes and prolifera-
limb ischemia or inoperable coronary artery tion rates comparable to larger, rapidly grow-
disease who undergo gene therapy with a ing tumors. This suggests that the difference
VEGF expression vector (43,44). Furthermore, between the two tumors is that in the prema-
human bone marrow stem cells were able to lignant lesion, the generation of new tumor
differentiate and contribute substantially to cells is balanced by tumor apoptosis (46). A
tumor vasculature in a mouse model (45). relatively small percentage of these lesions
Taken together, these studies suggest that bone undergo an "angiogenic switch," reflecting a
marrow-derived endothelial cells are likely to change in the net balance between angiogenic
make at least some contribution to tumor vas- stimulators and inhibitors (22). This appears
culature in humans, although the relative con- to be a critical step for sustained tumor growth

[Early stage I I Advanced stage ~


--...
Tumor growth

Angiogenic Vascular Angiogenic


sW~ch or lymphatic . _ _ _ _ _ sw~ch

. . . . . . . invasion

Avascular Vascularized Dormant Large tumor


Hyperplasia/dysplasia tumor micrometastases or metastasis
CIS

Therapeutic
goal: ---C-h-e-m-op-re-v-e-nt-lo-n---11--Ea-rIY-ln-te-rv-e-n-t-Io-n-: ------II--R -eg-re-s-s,-o-n-
Prevent growth and metastases

FIGURE 4.2. The role of angiogenesis in the progres- Vascularization also increases the likelihood of
sion of cancer. Premalignant lesions must acquire a metastatic spread. CIS, carcinoma in situ.
vascular supply to progress to macroscopic lesions.
4. Tumor Angiogenesis 45

and metastatic spread (Fig. 4.2). The angiogenic ducibly undergo a transition from normal islets
stimulus arising from preneoplastic lesions can to hyperplasia (60).A small percentage ofthese
be observed clinically: biopsies from patients hyperplastic lesions become angiogenic, leading
with preneoplastic bronchial, colonic, or cervi- to the development of tumors. Interestingly,
cal lesions ranging from hyperplasia and meta- different antiangiogenic agents (TNP-470,
plasia to carcinoma in situ often are associated endostatin, angiostatin, or the matrix metallo-
with increased microvessel density in the sur- proteinase inhibitor BB-94) had distinct activ-
rounding mucosa (47-49). The specific factors ity profiles in terms of their ability to prevent
controlling this switch have not been fully tumor formation (chemoprevention), slow the
elucidated and probably vary depending on the growth of small tumors (early intervention),
tumor type, but elevated levels of proangio- and regress established tumors (61). This high-
genic factors including VEGF, epidermal lights the importance of investigating the most
growth factor receptor (EGFR), and cyclooxy- appropriate applications for specific angiogen-
genase-2 (Cox-2) have been observed (50) in esis inhibitors.
preneoplastic lesions.
The majority of cancer deaths are caused by
metastases, not by the primary tumor. Evidence
from preclinical and clinical studies suggests Potential Advantages of
that the metastatic spread of cancer is also Antiangiogenic Therapy
angiogenesis dependent (1). For a tumor to
enter the circulation and successfully metasta- The approach of targeting the endothelium
size, it must first overcome several obstacles: it instead of, or in addition to, the tumor cell
must invade through the basement membrane, has a number of advantages over traditional
which is often fragmented and leaky in tumors; approaches focused on tumor cells alone.
survive in the circulation; arrest in the target Tumor endothelium appears to be distinct from
organ; and induce angiogenesis to resume normal endothelium at a molecular level, but
growth (51-55). In animal models, tumor cells endothelium from different tumor types appear
are rarely detectable in the circulation before to share tumor endothelium-specific markers
a primary tumor is vascularized, but can be (TEMs) (62) and other important characteris-
detected (at levels as high as 106 cells per gram tics, such as high proliferation rates. This raises
of tumor) after vascularization (56,57). Angio- the possibility that agents directed against
genic factors such as VEGF and bFGF increase tumor endothelium will be active for a broad
endothelial motility and induce production of range of tumor types, a hope that has been
proteinases that degrade the basement mem- supported by animal studies (63-65). It is
brane, further facilitating the entry of tumor important to note, however, that recent studies
cells into the circulation (56). Fortunately, the have highlighted the heterogeneity of different
vast majority of tumor cells that gain entry into endothelial beds in terms of markers and re-
the circulation do not develop into macroscopic sponsiveness to proangiogenic factors (66,67).
tumors. Such metastases may lack angiogenic It is likely that tumor endothelium from dif-
activity for a number of reasons, and therefore ferent tumor types will vary in their respon-
remain as a microscopic tumor of 100 to 200 ~m siveness to both angiogenic stimulators and
indefinitely (21,58). inhibitors.
Angiogenesis inhibitors, therefore, may offer Resistance to conventional chemotherapy
benefit when used for chemoprevention as well occurs, in large part, because genetic instability
as in treatment of early stage, occult metastatic, and high mutation rates inevitably lead to the
or advanced disease (59). This hypothesis has selection of resistant clones. Angiogenesis
been supported by a transgenic murine model inhibitors, by contrast, target activated endo-
of pancreatic islet carcinogenesis. In this model, thelial cells that are diploid and presumably
lesions in mice harboring an oncogene ex- genetically stable. This led to the prediction that
pressed in the p- islet cells of the pancreas repro- resistance will develop more slowly, or not at
46 IV. Heymach and I Folkman

all, to angiogenesis inhibitors. Consistent with conceptual framework for considering these
this hypothesis, resistance was not observed in drugs.
three different murine tumor models even after
six cycles of therapy with endostatin (68). It is
clear, however, that different tumor types do
Direct vs. Indirect Inhibitors
not respond identically to antiangiogenic It is useful to separate antiangiogenic agents
therapy and that genetic changes in the tumor into two broad categories: direct and indirect
can alter this responsiveness. For example, p53 inhibitors (Fig. 4.3) (73). Direct inhibitors, such
mutations are known to cause increased tumor as TNP-470, angiostatin, vitaxin, and others, act
angiogenesis through several mechanisms, directly on vascular endothelial cells to prevent
including VEGF upregulation and a reduction them from proliferating, migrating, or surviving
in the thrombospondin-l (TSP-l). p53 muta- in response to a spectrum of proangiogenic
tions also render tumor cells relatively resistant factors such as VEGF, bFGF, and PDGF (Table
to hypoxia-induced apoptosis. Not surprisingly, 4.2). Direct angiogenesis inhibitors should in
tumors derived from a p53-null human colo- theory be less prone to resistance because they
rectal cell line responded less well to antian- target genetically stable cells. Indirect inhi-
giogenic therapy than their p53 wild-type coun- bitors, on the other hand, block the expression
terparts, although their growth was still or activity of factors produced by tumor. For
inhibited (69). example, ZD1839 (Iressa) and Herceptin are
Antiangiogenic therapy differs from tradi- agents developed to inhibit the EGFR and
tional cytotoxic therapies in at least two other
important ways. First, because tumors have high
interstitial pressures, tumor blood flow is often
compromised, resulting in markedly diminished Indirect Direct
penetration of cytotoxic drugs into tumor Inhibitor lressa Endostatin
tissues (70). In contrast, antiangiogenic agents Inhibits synthesis Inhibits endothelial

1I
target endothelium that is in direct contact Mechanism by tumor cells cells from responding
of angiogenic to multiple angiogenic
with circulation and do not, therefore, need to

1
proteins: proteins, e.g., bFGF,
penetrate tissues to exert their effects. Second, bFGF, VEGF, VEGF, IL-B, PDGF.

the side-effect profiles of antiangiogenic agents .ooro~


have typically been quite favorable, and non- bFGF

~JJ
overlapping with chemotherapy. Two excep- VEGF
tions to this have been rare life-threatening side Tumor
effects such as hemoptysis (71) and blood clots
(72) that occurred primarily when VEGF . Endoi::;
pathway antagonists were given in combination cells
with chemotherapy. These side effects are dis-
cussed below. FIGURE 4.3. Direct vs. indirect angiogenesis inhi-
bitors. Direct angiogenesis inhibitors, such as endo-
statin, target tumor endothelial cells and prevent
Types of Angiogenesis Inhibitors them from responding to various tumor-derived
factors that stimulate angiogenesis. Indirect inhibitors
Research into the molecular mechanisms such as Iressa (ZD1839) target proteins that are
produced by tumor cells, such as VEGF, bFGF, and
underlying tumor angiogenesis has catalyzed a
TGF-a, or inhibit their receptors on endothelium.
rapid growth in the number of angiogenesis bFGF, basic fibroblast growth factor; VEGF, vascular
inhibitors developed over the past decade. An endothelial growth factor; TGF-a, transforming
exhaustive review of the large number of agents growth factor-a; IL-8, interleukin-8; PDGF, platelet-
entering clinical trials is beyond the scope of derived growth factor. (Reproduced with permission
this chapter. Instead, we will briefly review from Nature Reviews Cancer, Vol. 2, No. 10, pp.
several representative examples and discuss a 727-739, copyright 2002, Macmillan Magazines Ltd.)
4. Tumor Angiogenesis 47

TABLE 4.2. Examples of direct and indirect angiogenesis inhibitors in clinical trials.
Agent Proposed mechanism Clinical trials
Direct
Angiostatin Binds to ATP synthase and annexin II to inhibit EC proliferation Phase I
and migration
Combretastatin, ZD6126 Microtubule inhibitor (VTA) Phase I
Vitaxin, cilengitide Inhibit av~3 integrin Phase I
Endostatin Binds to several EC targets including as and av integrin, MMP2, Phase II
eNOS; prevents loss of adhesion to BM induced by VEGF and bFGF
NM-3 Inhibits EC proliferation, sprouting, and tube formation Phase I
Thalidomide Inhibits bFGF-angiogenesis; may also inhibit production ofVEGF, Phase II
bFGF,TNF-a
Indirect
ZD1839 Inhibits EGFR leading to decreased production of VEGF, bFGF, Phase III
TGF-a
Herceptin Inhibits Her-2, decreasing VEGF, TGF-a,Ang-l, etc Phase III
Celecoxib, rofecoxib Inhibit COX-2 Phase III

VTA, vascular targeting agent; mAb, monoclonal antibody; EC, endothelial cell; BM, basement membrane; VEGF, vas-
cular endothelial growth factor; bFGF, basic fibroblast growth factor; TNF, tumor necrosis factor; EGFR, epidermal growth
factor receptor; TGF, transforming growth factor; Ang, angiopoietin; Cox, cyclooxygenase.

Her-2 pathways on tumor cells. Recent studies gests a molecular basis for the longstanding
have established that blockade of these onco- clinical observation that removal of a primary
genic pathways also decreases the expression tumor often results in an acceleration in the
of proangiogenic factors such as VEGF, and growth of metastases. This led to an intensive
increases the effects of endogenous angiogene- search for additional tumor-derived antiangio-
sis inhibitors such as thrombospondin-1 (74). It genic agents.
is worth noting that these categories are not Angiostatin was initially purified from the
mutually exclusive; a number of drugs, such urine of mice bearing Lewis lung carcinoma, a
as interferon-a, have both direct effects on tumor that when implanted subcutaneously
endothelium and indirect effects on the pro- suppressed the growth of lung metastases via
duction of cytokines. inhibition of angiogenesis (79). It was found to
be a 38-kd internal fragment of plasminogen.
Tumors do not appear to directly produce
Endogenous Angiogenesis Inhibitors angiostatin, but rather they release proteases
In the early 1980s it was demonstrated that that cleave circulating plasminogen. In addition
interferon-a/~, a cytokine initially identified as to potent antiangiogenic activity in a variety
an antiviral agent, inhibited endothelial prolif- of tumor models, angiostatin potentiates the
eration (75). Other endogenous proteins such effects of radiation (80).
as platelet factor-4 (76,77) later were identified A similar strategy has been used to identify
to have antiangiogenic activity as well. Inter- a number of other inhibitors, including endo-
estingly, angiogenesis inhibitors also were pro- statin (81), tumstatin (82), canstatin (83), and
duced by tumors themselves. This led to the antiangiogenic antithrombin-III (84), which
proposal that the angiogenic phenotype of a are reviewed in detail elsewhere (1). In phase I
tumor was determined by the net balance of testing, endostatin was found to be safe, and
angiogenesis inhibitors and stimulators (22,78). evidence of antitumor and antiangiogenic activ-
While the production of angiogenesis inhibitors ity was observed (85,86). Angiostatin and endo-
by tumors may seem counterintuitive, it sug- statin are currently in phase II testing.
48 IV. Heymach and I Folkman

VEGF Pathway Antagonists molecule receptor tyrosine kinase inhibitors


(RTKIs). The newest generation of RTKIs, such
VEGF is expressed in the vast majority of as PTK787, ZD6474, and SU1l248, are oral
human tumors, and levels of VEGF protein drugs, an important advantage for therapies
have been correlated with poor prognosis in a that may need to be taken for sustained periods
variety of malignancies including lung, breast, of time (Table 4.3).
colon, and prostate cancer (87-90). VEGF In early clinical testing, some VEGF pathway
expression is regulated physiologically by inhibitors have shown promising activity both
hypoxia, and in tumors a number of oncogenes as a single agent and in combination with
have been found to regulate VEGF production chemotherapy. For example, in a randomized
including H-Ras, K-Ras, v-src, p53, Her-2, and phase II trial in patients with renal cell cancer,
bcl-2 (91,92). It performs a number of roles in bevacizumab significantly prolonged the time
the angiogenic process (Fig. 4.1), such as induc- for tumor progression compared to placebo,
ing endothelial migration and proliferation and and objective tumor shrinkage was observed
promoting ECs survival. In addition to these in 8% of patients (93). In another randomized
important roles, VEGF also mobilizes bone phase II trial, patients with colon cancer were
marrow-derived circulating ECs precursors, treated with a standard chemotherapy 5-fluo-
induces the expression of matrix metallopro- rouracil/leucovorin (5-FU/LV) regimen with or
teinases, and increases vascular permeability- without the addition of the VEGF monoclonal
it was, in fact, initially identified by Dvorak's antibody bevacizumab (BV) (94). Patients
group (20) in the 1980s as a vascular perme- receiving the combination containing low-dose
ability factor. Because of its widespread ex- BV had a higher objective response rate (40%
pression in tumors and its central role in vs. 17% for control arm), longer time to pro-
angiogenesis, VEGF has been the subject gression (5.2 vs. 9 months), and prolonged
of intense investigation, and a number of survival (13.8 vs. 21.5 months) than the control
inhibitors of the VEGF pathway are being arm (94). Phase III trials of bevacizumab are
tested in clinical trials (Table 4.3). ongoing.
The activity of VEGF appears to be primar- Two serious toxicities observed with VEGF
ily mediated by the tyrosine kinase receptor pathway inhibitors have been thrombosis
VEGFR-2, although other receptors such as (94,95), and hemoptysis (71), which occurred in
VEGFR-1, VEGFR-3, and neuropilin-1 likely the setting of rapid tumor necrosis. Both toxici-
play a role as well. Approaches to inhibiting ties are likely related to endothelial damage that
these pathways include antibodies to the ligand occurred when angiogenesis inhibitors were
(bevacizumab), antibodies to the receptor that given in combination with chemotherapy. This
blocks ligand binding (IMC-1Cll), and small highlights the need for further investigations

TABLE 4.3. Inhibitors of the VEGF pathway in clinical trials.


Agent Main target(s) Type Company Clinical trials
Bevacizumab VEGF mAb Genentech Phase III
SU5416 VEGFR-2 RTKI Sugen Phase III (discontinued)
SU6668 VEGFR-2, PDGF RTKI (oral) Sugen Phase II (discontinued)
SUl1248 VEGFR-2, PDGF RTKI (oral) Sugen Phase II
PTK787 VEGFR-2, PDGF RTKI (oral) Novartis Phase II
ZD6474 VEGFR-2, PDGF RTKI (oral) Astra Zeneca Phase II
IMC-lCll VEGFR-2 mAb Imclone Phase I
VEGF trap VEGF Soluble VEGFR-2 Regeneron Phase I
Angiozyme VEGFR-l ribozyme Ribozyme Pharmaceuticals Phase II

VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor; PDGF, platelet-derived growth factor; RTKI, recep-
tor tyrosine kinase inhibitor; mAb, monoclonal antibody.
4. Tumor Angiogenesis 49

into the optimal means for combining these in vivo, often exerting toxicity against endothe-
inhibitors with other treatment modalities. lial cells at concentrations one to two orders of
magnitude lower than those required for anti-
tumor effects (106,108,109).
Role of Angiogenesis in Other The antiangiogenic effects of cytotoxics
appear to be highly dependent on the dosing
Therapeutic Modalities: schedule. We developed a cyclophosphamide
"Incidental" Antiangiogenesis dosing schedule (170 mg/kg every 6 days)
that inhibited the growth of highly cyclo-
Most therapeutic approaches to fighting cancer phosphamide-resistant Lewis lung carcinoma
were developed with the goal of attacking the through the induction of endothelial apoptosis
cancer cell. Evidence suggests that many of (99). The combination of the angiogenesis
these treatment modalities actually exert their inhibitor TNP-470 with cyclophosphamide
effects, at least in part, though inhibition and administered by this schedule caused tumors
damage to tumor endothelial cells and vascula- to regress completely for longer than 600 days
ture (96-99). Examples of this are chemother- (Fig. 4.2). Other investigators have subse-
apy and radiofrequency ablation, discussed quently confirmed that frequent administration
below; today a number of "antiangiogenic" or of low doses of chemotherapeutics (i.e., vin-
"metronomic" chemotherapy regimens are blastine, cisplatin, or Adriamycin given every 3
undergoing clinical testing. Another example days) (92,108) can damage tumor endothelium
is radiation therapy. Endothelial damage and with limited host toxicity in murine models.
apoptosis induced by ionizing radiation appear Based on these observations such regimens are
to play a crucial role in both the antitumor often referred to as antiangiogenic or metro-
and toxic effects (100). Consistent with this nomic dosing. This effect is heightened by com-
hypothesis, antiangiogenic agents such as bination with VEGF antagonists such as the
angiostatin (80) and an antibody to VEGF act monoclonal VEGFR antibody DC101 (92,108).
synergistically with radiotherapy (101). Com- The rationale is that such frequent dosing
binations of antiangiogenic agents and radio- damages or inhibits rapidly dividing tumor
therapy are now undergoing clinical testing as endothelium with greater efficacy than con-
well. ventional regimens, without allowing time for
recovery.
VEGF is an important survival factor for
Antiangiogenic Effects endothelial cells damaged by chemotherapy
of Chemotherapy (110). Consistent with this observation, a
Chemotherapeutic agents traditionally have recombinant human monoclonal antibody
been selected based on their antitumor activity. against VEGF (rhuMab VEGF) augments
However, there is now substantial evidence that chemotherapy-induced endothelial cell death
endothelial cells also may be an important (106). Conventional regimens, by contrast, are
target for these drugs. Some cytotoxic agents typically based on the maximum tolerated dose
have been shown to exert antiangiogenic (MTD) with an interval for recovery from tox-
effects via their ability to induce endothelial icity and bone marrow suppression; this inter-
apoptosis or interfere with the key steps in the val appears to allow time for recovery of tumor
angiogenic cascade that include endothelial endothelial function as well (99). Because
proliferation, migration, or capillary tube for- endothelial cells, like other normal tissues such
mation (96). The chemotherapeutic agents that as gut epithelium, are more genetically stable,
interfere with microtubule formation such as they are less likely to acquire drug resistance
paclitaxel (102-104), docetaxel (105,106), and through genetic mutation. Therefore, they are
vinca alkaloids (92,107) appear to be particu- likely to remain sensitive to chemotherapy
larly potent antiangiogenic agents in vitro and regardless of whether the tumor is resistant.
50 IV. Heymach and I Folkman

Antivascular Effects of with intention of attacking cancer cells-such


Radiofrequency Ablation as chemotherapy, radiotherapy, and radiofre-
and Hyperthermia quency ablation-actually exert their effects, at
least in part, through effects on angiogenesis
Radiofrequency ablation provides a means to and tumor vasculature. Understanding the
induce local hyperthermia. There is a direct mechanisms by which different treatment
effect of heat on tumor cells, but in preclinical modalities exert their antitumor and antivascu-
models tumors may be cured despite the fact lar effects, therefore, is important for designing
that a significant percentage of tumor cells appropriate preclinical models, choosing other
remain viable at the end of therapy (111-114). agents to be combined with these modalities,
The effects of hyperthermia, therefore, extend and optimizing schedules of administration. In
beyond direct damage to tumor cells. Investiga- the near future, approaches aimed at targeting
tions into the changes in tumor microvascula- both tumor cells and tumor vasculature may
ture after hyperthermia have shown a brief lead to more effective-and less toxic-treat-
initial increase in blood flow followed by a pro- ments for cancer.
longed decrease and in some cases a cessation
of perfusion that leads to tissue hypoxia and
necrosis (98,113,115). These changes are accom-
panied by intravascular thrombosis, microvessel References
rupture, endothelial apoptosis, and an inhibi-
tion of angiogenesis (114,116). Proliferating 1. Folkman J, Kalluri R. Tumor angiogenesis. In:
Kufe DW, Pollock RE, Weichselbaum RR, Bast
endothelial cells in tumor vasculature, appear to
RC Jr, Holland JF, Frei III E, eds. Cancer
be more sensitive than endothelium from Medicine. 6th ed. Hamilton, Ontario: E.c.
normal tissues (98,117,118). This has led to Decker,2003.
the identification of tumor microvasculature as 2. Warren BA. The vascular morphology of
an important target for hyperthermia (97,98, tumors. In: Peterson HI, ed. Tumor Blood Cir-
119). culation: Angiogenesis, Vascular Morphology,
Tissue hypoxia induced by these antivascular and Blood Flow of Experimental Human
effects appears to induce the expression of pro- Tumors. Florida: CRC Press, 1979:1-47.
angiogenic factors such as VEGF and bFGF 3. Coman DR, Sheldon WE The significance of
(120). These factors are likely responsible for a hyperemia around tumor implants. Am J Pathol
1946;22:821-831.
compensatory wave of angiogenesis sometimes
4. Ide AG, Baker NH, Warren SL. Vasculariza-
observed after hyperthermia. With this in mind,
tion of the Brown-Pearce rabbit ephithelioma
it is not surprising that angiogenesis inhibitors transplant as seen in the transparent ear
such as TNP-470 augment the antitumor effi- chamber. AJR 1939;42:891-899.
cacy of hyperthermia (121-123). One would 5. Algire GH, Chalkely HW, Legallais FY, Park H.
expect that newer agents such as the VEGF Vascular reactions of normal and malignant
pathway antagonists (discussed above) could tumors in vivo: I. Vascular reactions of mice to
also potentiate the effects of hyperthermia, a wounds and to normal and neoplastic trans-
hypothesis that merits further investigation as plants. J Natl Cancer Inst 1945;6:73-85.
these agents move forward in clinical testing. 6. Folkman I Toward an understanding of angio-
genesis: search and discovery. Perspect BioI
Med 1985;29:10-36.
7. Folkman J, Cole P, Zimmerman S. Thmor behav-
Summary ior in isolated perfused organs: in vitro growth
and metastases of biopsy material in rabbit
Tumor growth is angiogenesis dependent, and thyroid and canine intestinal segment. Ann
currently various antiangiogenic agents are Surg 1966;164:491-502.
being tested in clinical trials for cancer and 8. Folkman J, Long DM, Becker FE Growth and
other diseases. Furthermore, it is now appreci- metastasis of tumor in organ culture. Cancer
ated that many therapies initially developed 1963;16:453-467.
4. Tumor Angiogenesis 51

9. Folkman 1. Tumor angiogenesis: therapeutic 23. Carmeliet P, Jain RK. Angiogenesis in cancer
implications. N Engl J Med 1971;285:1182- and other diseases. Nature 2000;407:249-257.
1186. 24. Liekens S, De Clercq E, Neyts 1. Angiogenesis:
10. Gimbrone MA, Leapman SB, Cotran RS, regulators and clinical applications. Biochem
Folkman 1. Tumor dormancy in vivo by preven- Pharmacol 2001;61:253-270.
tion of neovascularization. J Exp Med 1972; 25. Heymach JV. Angiogenesis and antiangiogenic
136:261-276. approaches to sarcomas. Curr Opin Onco12001;
11. Holmgren L, O'Reilly MS, Folkman 1. 13:261-269.
Dormancy of micrometastases: balanced pro- 26. Bergers G, Brekken R, McMahon G, et al.
liferation and apoptosis in the presence of Matrix metalloproteinase-9 triggers the angio-
angiogenesis suppression. Nat Med 1995;1: genic switch during carcinogenesis. Nat Cell
149-153. Bioi 2000;2:737-744.
12. Gimbrone MA Jr, Cotran RS, Folkman 1. 27. Moses MA, Wiederschain D, Loughlin KR,
Endothelial regeneration: studies with human Zurakowski D, Lamb CC, Freeman MR.
endothelial cells in culture. Ser Haematol1973; Increased incidence of matrix metallopro-
6:453-455. teinases in urine of cancer patients. Cancer Res
13. Jaffe EA, Nachman RL, Becker CG, Minick 1998;58:1395-1399.
CR. Culture of human endothelial cells derived 28. Maisonpierre PC, Suri C, Jones PF, et al.
from umbilical veins. Identification by morpho- Angiopoietin-2, a natural antagonist for Tie2
logic and immunologic criteria. J Clin Invest that disrupts in vivo angiogenesis. Science
1973;52:2745-2756. 1997;277:55-60.
14. Folkman J, Haudenschild CC, Zetter BR. Long- 29. Yancopoulos GD, Davis S, Gale NW, Rudge JS,
term culture of capillary endothelial cells. Proc Wiegand SJ, Holash 1. Vascular-specific growth
Nat! Acad Sci USA 1979;76:5217-5221. factors and blood vessel formation. Nature
15. Taylor S, Folkman 1. Protamine is an inhibitor 2000;407:242-248.
of angiogenesis. Nature 1982;297:307-312. 30. Suri C, Jones PF, Patan S, et al. Requisite role
16. Shing Y, Folkman J, Sullivan R, Butterfield C, of angiopoietin-1, a ligand for the TIE2 recep-
Murray J, Klagsbrun M. Heparin affinity: purifi- tor, during embryonic angiogenesis. Cell 1996;
cation of a tumor-derived capillary endothelial 87:1171-1180.
cell growth factor. Science 1984;223:1296-1299. 31. Gerber HP, Hillan KJ, Ryan AM, et al. VEGF
17. Klagsbrun M, Sasse J, Sullivan R, Smith JA. is required for growth and survival in neonatal
Human tumor cells synthesize an endothelial mice. Development 1999;126:1149-1159.
cell growth factor that is structurally related to 32. Senger DR, Perruzzi CA, Feder J, Dvorak
basic fibroblast growth factor. Proc Nat! Acad HE A highly conserved vascular permeability
Sci USA 1986;83:2448-2452. factor secreted by a variety of human and
18. Leung DW, Cachianes G, Kuang WJ, Goeddel rodent tumor cell lines. Cancer Res 1986;46:
DV, Ferrara N. Vascular endothelial growth 5629-5632.
factor is a secreted angiogenic mitogen. Science 33. Asahara T, Masuda H, Takahashi T, et al. Bone
1989;246:1306-1309. marrow origin of endothelial progenitor cells
19. Ferrara N, Henzel W1. Pituitary follicular cells responsible for postnatal vasculogenesis in
secrete a novel heparin-binding growth factor physiological and pathological neovasculariza-
specific for vascular endothelial cells. Biochem tion. Circ Res 1999;85:221-228.
Biophys Res Commun 1989;161:851-858. 34. Asahara T, Murohara T, Sullivan A, et al. Isola-
20. Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, tion of putative progenitor endothelial cells for
Harvey VS, Dvorak HE Tumor cells secrete angiogenesis. Science 1997;275:964-967.
a vascular permeability factor that promotes 35. Asahara T, Takahashi T, Masuda H, et al.
accumulation of ascites fluid. Science 1983;219: VEGF contributes to postnatal neovasculariza-
983-985. tion by mobilizing bone marrow-derived
21. Folkman 1. Angiogenesis in cancer, vascular, endothelial progenitor cells. EMBO J 1999;18:
rheumatoid and other disease. Nat Med 1995; 3964-3972.
1:27-31. 36. Lyden D, Young AZ, Zagzag D, et al. Id1 and
22. Hanahan D, Folkman 1. Patterns and emerging Id3 are required for neurogenesis, angiogenesis
mechanisms of the angiogenic switch during and vascularization of tumour xenografts.
tumorigenesis. Cell 1996;86:353-364. Nature 1999;401:670-677.
52 IV. Heymach and I Folkman

37. Lyden D, Hattori K, Dias S, et al. Impaired 49. Keith RL, Miller YE, Gemmill RM, et al.
recruitment of bone-marrow-derived endothe- Angiogenic squamous dysplasia in bronchi of
lial and hematopoietic precursor cells blocks individuals at high risk for lung cancer. Clin
tumor angiogenesis and growth. Nat Med 2001; Cancer Res 2000;6:1616--1625.
7:1194-1201. 50. Wolff H, Saukkonen K, Anttila S, Karjalainen
38. Rafii S, Meeus S, Dias S, et al. Contribution A, Vainio H, Ristimaki A. Expression of
of marrow-derived progenitors to vascular cyclooxygenase-2 in human lung carcinoma.
and cardiac regeneration. Semin Cell Dev Bioi Cancer Res 1998;58:4997-5001.
2002;13:61-67. 51. Fidler D, Gersten DM, Hart IR. The biology of
39. Peichev M, Naiyer AJ, Pereira D, et al. Expres- cancer invasion and metastasis. Adv Cancer
sion of VEGFR-2 and AC133 by circulating Res 1978;28:149-250.
human CD34(+) cells identifies a population 52. Nicolson G L. Organ specificity of tumor metas-
of functional endothelial precursors. Blood tasis: role of preferential adhesion, invasion and
2000;95:952-958. growth of malignant cells at specific secondary
40. Mancuso P, Burlini A, Pruneri G, Goldhirsch A, sites. Cancer Metastasis Rev 1988;7:143-188.
Martinelli G, Bertolini F. Resting and activated 53. Zebrowski BK, Yano S, Liu W, et al. Vascular
endothelial cells are increased in the peripheral endothelial growth factor levels and induction
blood of cancer patients. Blood 2001;97:3658- of permeability in malignant pleural effusions.
3661. Clin Cancer Res 1999;5:3364-3368.
41. Bertolini F, Mingrone W, Alietti A, et al. 54. Zetter BR. Angiogenesis and tumor metastasis.
Thalidomide in multiple myeloma, myelodys- Annu Rev Med 1998;49:407-424.
plastic syndromes and histiocytosis. Analysis of 55. Dvorak HF, Nagy JA, Dvorak JT, Dvorak AM.
clinical results and of surrogate angiogenesis Identification and characterization of the blood
markers. Ann OncoI2001;12:987-990. vessels of solid tumors that are leaky to circu-
42. Monestiroli S, Mancuso P, Burlini A, et al. lating macromolecules. Am J Pathol 1988;133:
Kinetics and viability of circulating endothelial 95-109.
cells as surrogate angiogenesis marker in an 56. Liotta LA, Tryggvason K, Garbisa S, Hart
animal model of human lymphoma. Cancer Res I, Foltz CM, Shafie S. Metastatic potential
2001;61:4341-4344. correlates with enzymatic degradation of
43. Kalka C, Tehrani H, Laudenberg B, et al. VEGF basement membrane collagen. Nature 1980;
gene transfer mobilizes endothelial progenitor 284:67-68.
cells in patients with inoperable coronary dis- 57. Chang YS, di Tomaso E, McDonald DM, Jones
ease. Ann Thorac Surg 2000;70:829-834. R, Jain RK, Munn LL. Mosaic blood vessels in
44. Kalka C, Masuda H, Takahashi T, et al. Vascu- tumors: frequency of cancer cells in contact
lar endothelial growth factor(165) gene trans- with flowing blood. Proc Nat! Acad Sci USA
fer augments circulating endothelial progenitor 2000;97:14608-14613.
cells in human subjects. Circ Res 2000;86: 58. Chambers AF. The metastatic process: basic
1198-1202. research and clinical implications. Oncol Res
45. Reyes M, Dudek A, Jahagirdar B, Koodie L, 1999;11:161-168.
Marker PH, Verfaillie CM. Origin of endothe- 59. Herbst RS, Hidalgo M, Pierson AS, Holden
lial progenitors in human postnatal bone SN, Bergen M, Eckhardt SG. Angiogenesis
marrow. J Clin Invest 2002;109:337-346. inhibitors in clinical development for lung
46. Udagawa T, Fernandez A, Achilles EG, cancer. Semin Oncol 2002;29:66--77.
Folkman J, D'Amato RI Persistence of micro- 60. Folkman J, Watson K, Ingber D, Hanahan D.
scopic human cancers in mice: alterations in the Induction of angiogenesis during the transition
angiogenic balance accompanies loss of tumor from hyperplasia to neoplasia. Nature 1989;339:
dormancy. FASEB J 2002;16:1361-1370. 58-61.
47. Fontanini G, Calcinai A, Boldrini L, et al. Mod- 61. Bergers G, Javaherian K, Lo KM, Folkman J,
ulation of neoangiogenesis in bronchial pre- Hanahan D. Effects of angiogenesis inhibitors
neoplastic lesions. Oncol Rep 1999;6:813-817. on multistage carcinogenesis in mice. Science
48. Smith-McCune KK, Weidner N. Demonstration 1999;284:808-812.
and characterization of the angiogenic proper- 62. St Croix B, Rago C, Velculescu V, et al.
ties of cervical dysplasia. Cancer Res 1994;54: Genes expressed in human tumor endothelium.
800-804. Science 2000;289~1197-1202.
4. Tumor Angiogenesis 53

63. Wedge SR, Ogilvie DJ, Dukes M, et a1. ZD6474 a selective epidermal growth factor receptor
inhibits vascular endothelial growth factor tyrosine kinase inhibitor. Clin Cancer Res 2001;
signaling, angiogenesis, and tumor growth 7:1459-1465.
following oral administration. Cancer Res 2002; 75. Brouty-Boye D, Zetter BR. Inhibition of cell
62:4645-4655. motility by interferon. Science 1980;208:516-
64. Fong TA, Shawver LK, Sun L, et a1. SU5416 518.
is a potent and selective inhibitor of the vas- 76. Maione TE, Gray GS, Petro J, et a1. Inhibition
cular endothelial growth factor receptor (Flk- of angiogenesis by recombinant human platelet
I/KDR) that inhibits tyrosine kinase catalysis, factor-4 and related peptides. Science 1990;247:
tumor vascularization, and growth of multiple 77-79.
tumor types. Cancer Res 1999;59:99-106. 77. Rastinejad F, Polverini PJ, Bouck NP. Regula-
65. Wood JM, Bold G, Buchdunger E, et a1. tion of the activity of a new inhibitor of angio-
PTK787/ZK 222584, a novel and potent genesis by a cancer suppressor gene. Cell 1989;
inhibitor of vascular endothelial growth factor 56:345-355.
receptor tyrosine kinases, impairs vascular 78. Bouck N. Tumor angiogenesis: the role of onco-
endothelial growth factor-induced responses genes and tumor suppressor genes. Cancer
and tumor growth after oral administration. Cells 1990;2:179-185.
Cancer Res 2000;60:2178-2189. 79. O'Reilly MS, Holmgren L, Shing Y, et a1. Angio-
66. Lin R, LeCouter J, Kowalski J, Ferrara N. statin: a novel angiogenesis inhibitor that medi-
Characterization of endocrine gland-derived ates the suppression of metastases by a Lewis
vascular endothelial growth factor signaling in lung carcinoma. Cell 1994;79:315-328.
adrenal cortex capillary endothelial cells. J BioI 80. Mauceri HJ, Hanna NN, Beckett MA, et a1.
Chern 2002;277:8724-8729. Combined effects of angiostatin and ionizing
67. Arap W, Kolonin MG, Trepel M, et a1. Steps radiation in antitumour therapy. Nature 1998;
toward mapping the human vasculature by 394:287-291.
phage display. Nat Med 2002;8:121-127. 81. O'Reilly MS, Boehm T, Shing Y, et a1. Endo-
68. Boehm T, Folkman J, Browder T, O'Reilly statin: an endogenous inhibitor of angiogenesis
MS. Antiangiogenic therapy of experimental and tumor growth. Cell 1997;88:277-285.
cancer does not induce acquired drug resis- 82. Maeshima Y, Colorado PC, Torre A, et a1. Dis-
tance. Nature 1997;390:404-407. tinct antitumor properties of a type IV collagen
69. Yu JL, Rak JW, Coomber BL, Hicklin DJ, domain derived from basement membrane.
Kerbel RS. Effect of p53 status on tumor J BioI Chern 2000;275:21340-21348.
response to antiangiogenic therapy. Science 83. Kamphaus GD, Colorado PC, Panka DJ, et a1.
2002;295: 1526-1528. Canstatin, a novel matrix-derived inhibitor of
70. Jain RK. Normalizing tumor vasculature with angiogenesis and tumor growth. J Bioi Chern
anti-angiogenic therapy: a new paradigm for 2000;275:1209-1215.
combination therapy. Nat Med 2001;7:987-989. 84. O'Reilly MS, Pirie-Shepherd S, Lane WS,
71. DeVore RF, Fehrenbacher L, Herbst RS, et a1. Folkman 1. Antiangiogenic activity of the
A randomized phase II trial comparing cleaved conformation of the serpin antithrom-
Rhumab VEGF (recombinant humanized bin. Science 1999;285:1926-1928.
monoclonal antibody to vascular endothelial 85. Herbst RS, Tran HT, Mullani NA, et a1. Phase I
cell growth factor) plus carboplatin/paclitaxel clinical trial of recombinant human endostatin
(CP) to CP alone in patients with stage IIIB/IV (rHE) in patients (Pts) with solid tumors: phar-
NSCLC. Proc Am Soc Clin OncoI2000;19:1896. macokinetic (PK), safety and efficacy analysis
72. Kuenen BC, Rosen L, Smit EF, et a1. Dose- using surrogate endpoints of tissue and radio-
finding and pharmacokinetic study of cisplatin, logic response. Proc Am Soc Clin Oncol 2001;
gemcitabine, and SU5416 in patients with solid 20:9.
tumors. J Clin Oncol 2002;20:1657-1667. 86. Eder JP Jr, Supko JG, Clark JW, et al. Phase I
73. Kerbel R, Folkman J. Clinical translation of clinical trial of recombinant human endostatin
angiogenesis inhibitors. Nature Rev 2002;2: administered as a short intravenous infusion
727-739. repeated daily. J Clin Oncol 2002;20:3772-3784.
74. Ciardiello F, Caputo R, Bianco R, et a1. Inhibi- 87. Fontanini G, Vignati S, Boldrini L, et a1. Vascu-
tion of growth factor production and angiogen- lar endothelial growth factor is associated with
esis in human cancer cells by ZD1839 (Iressa), neovascularization and influences progression
54 1.v. Heymach and 1. Folkman

of non-small cell lung carcinoma. Clin Cancer 98. Reinhold HS, Endrich B. Tumour microcircula-
Res 1997;3:861-865. tion as a target for hyperthermia. Int J Hyper-
88. Yuan A, Yu CJ, Chen WJ, et al. Correlation thermia 1986;2:111-137.
of total VEGF mRNA and protein expression 99. Browder T, Butterfield CE, Kraling BM, et al.
with histologic type, tumor angiogenesis, Antiangiogenic scheduling of chemotherapy
patient survival and timing of relapse in non- improves efficacy against experimental drug-
small-cell lung cancer. Int J Cancer 2000;89: resistant cancer. Cancer Res 2000;60:1878-
475-483. 1886.
89. George DJ, Halabi S, Shepard TF, et al. 100. Paris F, Fuks Z, Kang A, et al. Endothelial
Prognostic significance of plasma vascular endo- apoptosis as the primary lesion initiating
thelial growth factor levels in patients with intestinal radiation damage in mice. Science
hormone-refractory prostate cancer treated on 2001;293:293-297.
Cancer and Leukemia Group B 9480. Clin 101. Gorski DH, Beckett MA, Jaskowiak NT, et al.
Cancer Res 2001;7:1932-1936. Blockage of the vascular endothelial growth
90. Gasparini G. Prognostic value of vascular factor stress response increases the antitumor
endothelial growth factor in breast cancer. effects of ionizing radiation. Cancer Res 1999;
Oncologist 2oo0;5(suppl1):37-44. 59:3374--3378.
91. Rak J, Mitsuhashi Y, Bayko L, et al. Mutant ras 102. Belotti D, Vergani V, Drudis T, et al. The
oncogenes upregulate VEGFNPF expression: microtubule-affecting drug paclitaxel has
implications for induction and inhibition of antiangiogenic activity. Clin Cancer Res 1996;
tumor angiogenesis. Cancer Res 1995;55:4575- 2:1843-1849.
4580. 103. Klauber N, Parangi S, Flynn E, Hamel E,
92. Kerbel RS, Viloria-Petit A, Klement G, Rak 1. D'Amato RI. Inhibition of angiogenesis and
'Accidental' anti-angiogenic drugs. anti- breast cancer in mice by the microtubule
oncogene directed signal transduction inhibi- inhibitors 2-methoxyestradiol and taxol.
tors and conventional chemotherapeutic agents Cancer Res 1997;57:81-86.
as examples. Eur J Cancer 2000;36:1248- 104. Lau DH, Xue L, Young D, Burke PA, Cheung
1257. AT. Paclitaxel (Taxol): an inhibitor of angio-
93. Yang JC, Haworth L, Steinberg SM, Rosenberg genesis in a highly vascularized transgenic
SA, Novotny W. A randomized double-blind breast cancer. Cancer Biother Radiopharm
placebo-controlled trial of bevacizumab (anti- 1999;14:31-36.
VEGF antibody) demonstrating a prolonga- 105. Vacca A, Ribatti D, Iurlaro M, et al. Docetaxel
tion in time to progression in patients with versus Paclitaxel for antiangiogenesis. J Hema-
metastatic renal cancer. NEJM 2003;349(5): tother Stem Cell Res 2002;11:103-118.
427-434. 106. Sweeney CJ, Miller KD, Sissons SE, et al. The
94. Kabbinavar F, Hurwitz HI, Fehrenbacher L, antiangiogenic property of docetaxel is syner-
et al. Phase II, randomized trial comparing gistic with a recombinant humanized mono-
bevacizumab plus fluorouracil (FU)/leucovorin clonal antibody against vascular endothelial
(LV) with FU/LV alone in patients with growth factor or 2-methoxyestradiol but antag-
metastatic colorectal cancer. J Clin Oncol 2003; onized by endothelial growth factors. Cancer
21:6D-65. Res 2001;61:3369-3372.
95. Kuenen BC, Levi M, Meijers JC, et al. Analysis 107. Vacca A, Iurlaro M, Ribatti D, et al. Antiangio-
of coagulation cascade and endothelial cell acti- genesis is produced by nontoxic doses of vin-
vation during inhibition of vascular endothelial blastine. Blood 1999;94:4143-4155.
growth factor/vascular endothelial growth 108. Klement G, Huang P, Mayer B, et al. Differ-
factor receptor pathway in cancer patients. ences in therapeutic indexes of combination
Arterioscler Thromb Vasc BioI 2002;22:1500- metronomic chemotherapy and an anti-
1505. VEGFR-2 antibody in multidrug-resistant
96. Miller KD, Sweeney CJ, Sledge GW Jr. Redefin- human breast cancer xenografts. Clin Cancer
ing the target: chemotherapeutics as antiangio- Res 2002;8:221-232.
genics. J Clin OncoI2001;19:1195-1206. 109. Wang J, Lou P, Lesniewski R, Henkin 1. Pacli-
97. Denekamp 1. Inadequate vasculature in solid taxel at ultra low concentrations inhibits angio-
tumours: consequences for cancer research genesis without affecting cellular microtubule
strategies. BJR SuppI1992;24:111-117. assembly. Anticancer Drugs 2003;14:13-19.
4. Tumor Angiogenesis 55

110. Tran J, Master Z, Yu JL, Rak J, Dumont DJ, vascular injury in normal and neoplastic tissue.
Kerbel RS. A role for surviving in chemoresis- Cancer 1985;56:991-1000.
tance of endothelial cells mediated by VEGF. 118. Song CWO Effect of hyperthermia on vascular
Proc Nat! Acad Sci USA 2002;99:4349-4354. functions of normal tissues and experimental
111. Song CW, Kang MS, Rhee JG, Levitt SH. Effect tumors; brief communication. J Nat! Cancer
of hyperthermia on vascular function in normal Inst 1978;60:711-713.
and neoplastic tissues. Ann NY Acad Sci 1980; 119. Fajardo LF, Prionas So. Endothelial cells
335:35-47. and hyperthermia. Int J Hyperthermia 1994;10:
112. Song CW, Kang MS, Rhee JG, Levitt SH. Vas- 347-353.
cular damage and delayed cell death in tumours 120. Kanamori S, Nishimura Y, Okuno Y, Horii N,
after hyperthermia. Br J Cancer 1980;41:309- Saga T, Hiraoka M. Induction of vascular
312. endothelial growth factor (VEGF) by hyper-
113. Fajardo LF, Egbert B, Marmor J, Hahn GM. thermia and/or an angiogenesis inhibitor. Int J
Effects of hyperthermia in a malignant tumor. Hyperthermia 1999;15:267-278.
Cancer 1980;45:613--623. 121. Ikeda S, Akagi K, Shiraishi T, Tanaka Y.
114. Fajardo LF, Prionas SD, Kowalski J, Kwan HH. Enhancement of the effect of an angiogenesis
Hyperthermia inhibits angiogenesis. Radiat inhibitor on murine tumors by hyperthermia.
Res 1988;114:297-306. Oncol Rep 1998;5:181-184.
115. Eikesdal Hp, Bjorkhaug ST, Dahl 0. Hyper- 122. Nishimura Y, Murata R, Hiraoka M. Combined
thermia exhibits anti-vascular activity in the s.c. effects of an angiogenesis inhibitor (TNP-470)
BT4An rat glioma: lack of interaction with the and hyperthermia. Br J Cancer 1996;73:270-
angiogenesis inhibitor batimastat. Int J Hyper- 274.
thermia 2002;18:141-152. 123. Yano T, Tanase M, Watanabe A, et al.
116. Nishimura Y, Hiraoka M, Jo S, et al. Microan- Enhancement effect of an anti-angiogenic
giographic and histologic analysis of the effects agent, TNP-470, on hyperthermia-induced
of hyperthermia on murine tumor vasculature. growth suppression of human esophageal and
Int J Radiat Oncol BioI Phys 1988;15:411-420. gastric cancers transplantable to nude mice.
117. Badylak SF, Babbs CF, Skojac TM, Voorhees Anticancer Res 1995;15:1355-1358.
WD, Richardson RC. Hyperthermia-induced
Section II
Operations for Tumor Ablation
5
Image-Guided Tumor Ablation:
How to Build a Practice
Gary Onik

Integrating the occasional tumor ablation pro- When I first started treating prostate cancer, I
cedure into a thriving radiology practice is not had an enormous amount of information to
a particularly difficult job. Patient preoperative digest about the treatment of the disease. Since
workup, billing, and patient follow-up need not the most useful prostate imaging modality,
change from the usual handling of other inter- transrectal ultrasound, is generally carried out
ventianal radiology or cross-sectional imaging by urologists, I, as with most radiologists, had
patients. If the intent, however, is to build a little experience with this organ or modality.
large, thriving tumor ablation service, many Except for reading the occasional prostate
issues need to be addressed if the venture is to magnetic resonance imaging (MRI) or perusing
be successful, without placing undue time and a bone scan to rule out prostate metastasis,
financial burden on a radiology practice. My radiologists have few instances in which knowl-
practice is image-guided tumor ablation exclu- edge of the clinical and pathologic aspects of
sively, and therefore this chapter focuses on prostate cancer is relevant.
problems and pitfalls associated with building a It was not until I started actively managing
successful tumor ablation practice. these patients myself that I found it necessary
Many concepts in this chapter are a work in to become conversant with the patient selection
progress, since many difficult issues have yet to for the various prostate cancer treatments,
be satisfactorily resolved. There are two major the significance of both staging and pathologic
subject areas to be addressed when growing grading of prostate cancer, the benchmark suc-
such an ablation practice: acquiring the pa- cess rates, and the possible outcomes and com-
tients, and receiving adequate reimbursement plications of alternative procedures. Without
to make the effort worthwhile. It is truly prob- that knowledge it was impossible to give ade-
lematic to lose money on the treatment of each quate information to patients as to how this
patient, while trying to make up for the short- new ablation treatment fit into treatment
fall in income with the increased volume as the options. In addition, since there was now com-
ablation practice grows. We will first discuss the petition from other physicians practicing more
ins and outs of building the ablation practice, traditional prostate cancer therapies, I had to
and then address the more difficult aspect of defend the right to treat these patients in
adequate reimbursement. one forum or another (the institutional review
board [IRB] or the committee overseeing priv-
Getting Started ileges). Only by having a firm grasp of prostate
cancer was I able to expose the weaknesses in
The most basic step in starting an ablation the arguments of those trying to defend their
practice is to become an expert in the overall turf. Secondarily, my ability to converse in the
management of the cancer you will be treating. "language" of prostate cancer markedly in-

59
60 G. Onik

creased my credibility with referring primary best overall outcome can be obtained by
physicians and sophisticated patients, who treating these unsuspected lesions as early as
are extremely well informed through the possible before they appear on nonoperative
Internet. cross-sectional imaging.
Along with the need to become broadly clin- Another factor is that at least one recent
ically proficient, a commitment to establish an study has shown a survival advantage in treat-
"ablation clinic" almost certainly will have to ing hepatic resection patients with adjuvant
be made. In this clinic, patients will be seen for long-term intraarterial infusional chemother-
their preprocedure evaluation, given informed apy delivered by implantable pump combined
consent forms, and perhaps even undergoing a with systemic chemotherapy (1). We found it a
problem-directed physical exam prior to sched- compelling argument that the same advantage
uling the procedure. Doing a procedure on the would be realized in ablation patients as well.
same day as seeing the patient for the first time Since almost all of our patients are going to
should be avoided, since it does not allow have at least a minilaparotomy for the pump
development of an adequate physician- placement, it makes sense to do an intraopera-
patient relationship. This relationship is critical tive ultrasound and ablation at the same time.
to help defend malpractice suits when an In practicality, in our team approach, after the
inevitable ablation complication occurs. laparotomy is performed, the radiologist is
A decision also has to be made at the outset called in to carry out the intraoperative ultra-
as to whether to practice alone or establish a sound. Decisions are made jointly between sur-
team approach with the other specialties at geons and radiologists as to which lesions will
your institution. Such an approach would be be resected and which will be ablated. The
akin to the Miami Vascular Institute concept so pump is placed, any lesions to be resected are
successfully instituted by Dr. Barry Katzen. carried out first, since radiofrequency ablation
Establishing, for instance, a liver cancer insti- (RFA) or cryosurgery can increase clotting
tute in which the medical and surgical oncolo- times and decrease platelet counts, and then the
gists are represented has certain advantages, radiologist is called back to do the ablation. In
and has the potential to provide more coordi- other words, if a real commitment is going to
nated and therefore superior patient care. Such made to a liver ablation program, it is almost
an approach can simplify political issues, widen inevitable that radiologists will have to do some
the scope of the patients to be treated (doing work intraoperatively and interact on a coordi-
intraoperative ablations with surgical oncolo- nated basis with both medical and surgical
gists), and provide collegial backup for the oncologists.
complications that may occur. Take for instance Another decision that needs to be dealt with
the treatment of patients with liver metastasis when starting a program is the choice of abla-
from colon carcinoma. In our experience, an tion equipment. Since each ablation method
overwhelming number of patients in this situa- has its own advantages and disadvantages,
tion will not be amenable to a percutaneous having both RFA and cryosurgery available
ablation therapy in the radiology department. can be advantageous. There is much hype and
Limiting factors to treating patients with a per- misinformation in the marketplace as to lesion
cutaneous approach include too many lesions, size, shape, etc. created by the various RF
lesions that are too large, and a lesion adjacent systems. At the very least, a side-by-side com-
to a structure that is at risk for thermal injury, parison of the three systems that are currently
such as bowel. Many of these patients, however, available should be carried out. Whenever I am
can be treated in an intraoperative environ- evaluating new ablation technology, I use a
ment that allows larger ablations or even resec- large rump roast as my experimental model.
tion combined with ablation. A good case can The relative size and shape, as well as time
be made that since intraoperative (maybe even needed to make the lesion, will all become
laparoscopic) ultrasound is still the most sensi- clear as the lesions are created with the various
tive means to find lesions in these patients, the systems.
5. Image-Guided Tumor Ablation: How to Build a Practice 61

The final consideration when starting the referrals for ablation, particularly when clini-
program is data collection. Clinical data are cians learn that ablation therapy does not
the currency by which the program will grow preclude traditional therapy, but rather often
and gain legitimacy. As soon as the program is augments it.
started, therefore, a database should be initi- Initially, when starting the program other
ated in which the patient's clinical, procedural, noninterventional members of the radiology
and postprocedure data are entered prospec- department should be informed of the cases
tively. With such a database, valuable informa- that would be amenable for treatment. The
tion will be sure to be collected on every "ablationist" can then contact the referring
patient, with the result that patients are far less physician to explain that a new treatment
likely to be lost to follow-up, and trends that modality is available. This is the single most
affect patient care such as local recurrence rates effective means of initially gaining access to the
will be tracked without fail. Such a database local patient population.
also allows new patients to know when the test
results can be expected. If the results are avail-
able sooner than those reported by more tradi-
The Internet
tional treatment, your practice will be in a good If the most important mantra for the real estate
position in competing for patients. Also, industry is "location, location, location", then
without this prospective data collection, it is the equivalent for building an ablation practice
much less likely that your data will be pub- is "the Internet, the Internet, the Internet".
lished, since nothing cools the ardor for writing The Internet is revolutionizing the delivery of
like the thought of poring though a huge stack medical care. For the first time, patients have
of charts to extract data. ready access to health information without
the physician being the filter. It is therefore
much more likely that a patient will learn of
Growing the Practice an emerging treatment. Coupled with a growing
skepticism of the medical profession and a
The traditional model of growing a specialty growing acceptance of alternative medical
practice by patient referral from local prac- treatments, it follows that patients can research
ticing physicians is undergoing a revolution, their own illness through the Internet and
as patients gain more access to information choose newer treatments despite a negative
through the Internet, patient support groups, bias of their treating physician. The impact that
and the media. More and more patients are this can have on an ablation practice can be
supplementing the information their physician enormous. About 75% of patients that I treat
gives them with information from these have learned of tumor ablation through the
sources, and are taking more responsibility for Internet, and have heard about my practice on
the ultimate treatment decisions. The greater the Internet. But on the downside, clinicians
acceptance of alternative medicine also has become more motivated to improve the deliv-
made patients more receptive to newer treat- ery of patient care because they realize that a
ments if they are properly informed. A physi- disgruntled patient can reach the whole world
cian wishing to build a tumor ablation practice by posting on patient-oriented Web sites and
beyond the occasional liver ablation will even- disease-specific patient chat rooms negative
tually have to address the issue of direct patient comments about your lack of expertise, poor
marketing. bedside manner, and bad breath.
Don't skimp on the development of an
engaging and informative Web site; this is a job
Sources of Local Patients for a professional. Usually, your institution can
As already stated, a team approach that pro- be counted on to contribute significantly to this
vides comprehensive cancer care is a founda- endeavor if its name is prominently displayed
tion for building the practice and for attracting and incorporated into the site. Once the Web
62 G. Onik

site is up and running, it is important to hire a you desire national media exposure, the only
professional to maintain it and to continually reliable avenue is through publication of an
improve its visibility among the various search article in a media-recognized journal (JAMA,
engines. How search engines rank their various New England Journal of Medicine, or a subspe-
results is critical. The Web site consultant cialty journal) or presentation of an abstract
should be included in the process of choosing at a major meeting such as the Radiological
the site name and Internet address, as these are Society of North America (RSNA). Some of the
important for a successful ranking. An ongoing best media exposure has occurred through the
fee will have to be paid to this consultant to excellent work of the RSNA public relations
maintain the Web site, explore links to advan- staff, which organizes press releases and press
tageous sites, continually monitor the Web site conferences based on abstracts presented at the
activity, and make adjustments to maintain meeting.
your ranking. Media exposure can be a double-edged
We get a monthly report of Internet traffic at sword, however, particularly if the topic is con-
our site and on our ranking, based on various sidered controversial and has opponents in the
key words for the different search engines. One particular subspecialty of the traditional treat-
can fall from a number one or two ranking in a ment. Any major national program will seek
key area on a major search engine to number out and present the negative view of your treat-
ten within less than a week. On a practical level, ment from a well-known "expert" in the field.
we all save time and money by referring inter- This expert usually has a vested interest in
ested patients for more information to the Web seeing that your new ablation procedure is con-
site first, rather than sending out expensive sidered about as credible as reading the bumps
brochures. On a cost versus benefit basis, a good on a patient's head to diagnosis prostate cancer,
Web site has great value. and will undoubtedly cast a negative light on
this new major treatment advance. The worst
media experience I had was engineered by a
The Media prominent orthopedic surgeon who was vehe-
As someone who has the dubious distinction mently opposed to a new procedure I had
of having had two articles written about his invented that was capturing a lot of attention at
work in the tabloid press, I feel like an expert the time (percutaneous discectomy). Unfortu-
on the positive and negative aspect of media nately, he had a friend who was a producer at a
exposure. Just to set the record straight, the major television network whom he was able to
standard of journalism associated with medical convince that percutaneous discectomy was an
reporting in the tabloid paper was among the untested procedure that was being foisted on
highest I have encountered, with all facts being an unsuspecting public for purely financial gain.
checked and the final article being read back to This was despite its approval from the National
me before publication to verify its accuracy. Blue Cross/Blue Shield Technology Assessment
This is a standard I have found lacking in other Committee, and the rigorous review it had
more prestigious outlets. undergone. No amount of contrary information
At the very least, the local media should be we provided was going to change the slant of
contacted (with the help of the hospital public that story. On the whole, though, national
relations department), announcing the start of media exposure can have a dramatic positive
the ablation program and the implications impact on an ablation practice, and should be
for medical care in the community. Often, the pursued if at all feasible.
company that sells the ablation equipment will There is another strategy for large-scale
have a media program that helps gain local nationwide exposure. Most local news stations
exposure. I was chagrined recently to find com- don't have the time, expertise, or money to
petitors across town being hailed by the local develop medical news content. Therefore, they
newspaper as "pioneers in this new cancer are amenable to using any credible content
treatment", after they treated just one case. If that is provided to them in a form that can be
5. Image-Guided Tumor Ablation: How to Build a Practice 63

tailored to their needs. Television production by Medicare has set reimbursement so low as
companies will produce a news segment in its to make performance of the procedure barely
appropriate form to be sent to local news affil- viable financially, if at all. Many patients would
iates across the country. The local affiliates then gladly pay cash to be treated, but federal law
take the footage provided and add their own prohibits this approach.
commentary. This is an expensive option, but it
usually produces significant results.
Conclusion
Payment
Image-guided tumor ablation is going to have a
Dealing with Current Procedural Terminology major impact on the practice of oncology in the
(CPT) codes are beyond the scope of this future. As with any new image-guided treat-
chapter, but proper planning can have a major ment, radiologists will have to rapidly enter the
impact on the financial viability of an ablation field to prevent it from being usurped by other
program. The reimbursement provided by stan- specialties. As already stated, a cooperative
dard insurance contracts for ablation proce- approach with the other oncology subspecial-
dures is poor. If balance billing is precluded ties can offer advantages, but radiologists
by the radiology group's contract, then certain should be aware that ultimately an independent
ablation procedures may become financially flow of patients directly to the practice is the
unfeasible. If an attempt to grow an ablation only assurance of a viable long-term program.
program is being planned, an effort should be
made to carve tumor ablation out of the con-
tract, to at least allow balance billing. Ironically, Reference
the financial environment is often much better 1. Kemeny N, Huang Y, Cohen AM, et al. Hepatic
when cash payment is possible, before the arterial infusion of chemotherapy after resection
approval of a procedure with a CPT code. For of hepatic metastases from colorectal cancer.
instance, the approval of prostate cryosurgery N Engl J Med 1999;341(27):2039-2047.
6
Anesthesia for Ablation
John A. Fox and Alan M. Harvey

This chapter provides a short primer on anes- First, patients with more extensive tumors
thesiology for clinicians who are involved in the can be selected for treatment. During tumor
care of tumor ablation patients, but who have ablation using computed tomography (CT) or
not been trained in one of the surgical subspe- MRI guidance, the interventional radiologist is
cialties. Anesthesiologists, in the role of pain involved in a myriad of tasks to localize the
treatment specialists, have long been asked to lesion, accurately place the device delivering
care for patients with metastatic tumors; the role the treatment, and then monitor the treatment
of anesthesiologists in tumor ablation therapy results. Each of these tasks requires numerous
represents a continuation of a recent trend that careful steps often coupled with multiple scans;
has seen anesthesiologists perform their con- the attention of the interventionalist is, of
ventional services (i.e., to make the patient necessity, on these steps and scans. In pa-
insensible to pain) in areas outside of the oper- tients with significant cardiopulmonary disease,
ating room. Anesthesiologists have expanded it may be inappropriate to assign the task of
their roles in the past several years in angio- vital sign monitoring to nonphysician person-
graphy, endoscopy, and magnetic resonance nel. Additionally, should any complication
imaging (MRI)-guided surgical suites, in which occur, all anesthesiologists, who, by the nature
they interact with physician and nonphysician of their subspecialty, are already experts in the
personnel who are as unfamiliar with what the "airway" section of Advanced Cardiac Life
anesthesiologist needs to deliver patient care as Support, would already be present, on site, to
the anesthesiologist is with the needs of the team treat airway events and to assist in the treat-
members in these non-operating-room settings. ment of cardiac events.
Many other chapters in this book discuss the Second, anesthesiology has made a point of
goals of the tumor ablation team. This chapter emphasizing patient safety in its practice.
discusses the goals of the anesthesiologist. Thus, Accordingly, it has advocated the use of phys-
this chapter will be as instructive for interven- iologic monitors and practice standards, and
tional radiologists as the remaining chapters it encourages discussing ways of improving pa-
were for us, the anesthesiologists. tient outcomes. These attributes can serve as a
guide for the interventional radiologist when
initiating and evaluating the safety and results
Advantages of Anesthesiology of a tumor ablation program.
Involvement Third, the presence of an anesthesiologist in
the care of the tumor ablation patient ensures
The role of the anesthesiologist in tumor abla- that the patient will have minimal pain during
tion therapy is to facilitate patient safety and the procedure. In most hospitals, anesthesiolo-
patient satisfaction. gists are the only clinicians who can supersede

64
6. Anesthesia for Ablation 65

the sedative and narcotic dosage outlined in plan may be canceled for the day or be signifi-
conscious sedation protocols and have the cantly delayed. If the care is to be in the CT or
familiarity with the pharmacology of these MRI suite, this could lead to long waits by other
medications to adjust the dosage according to patients needing the scanners, the waste of
the patient's prior medical condition. Specifi- valuable technician time, and other costly
cally, tumor patients will often be on other med- inefficiencies.
ications that could increase their resistance to Additionally an anesthesiology department
standard doses of pain medications. In patients needs to allocate an individual to administer the
who have large tumors that would require mul- anesthesia during the ablation procedure. Of
tiple treatments, this could lead to increased necessity this requires that the individual not be
patient satisfaction by minimizing the anxiety of available for the care of other patients in the
the patient about repeated visits and treatments. practice, and, depending on the clinical load of
Fourth, tumor ablation therapy outside of the hospital, may require recruitment and hiring
the operating room is, in most institutions, a new of additional anesthesiology practitioners.
program involving new equipment and new
treatment plans. Inherent in new programs are
unforeseen equipment and patient issues that Anesthesia Standards
may entail prolonged periods of time in which
the patient must lie still in the CT or MRI tube. Preoperative Visit
Active participation by an anesthesiologist
The preoperative visit serves several purposes
would allow the treatment plan to be carried out
and is the standard of care in the preoperative
in a safe, painless, and expeditious way.
preparation of the patient (2). On either the
day of the procedure or, ideally, several days
before the anticipated procedure, the anesthe-
Disadvantages of Anesthesiology sia options are explained to the patient and
consent for the anesthesia is signed. A history
Involvement is taken and a physical examination is per-
formed, both focusing on the anesthesia prior-
Involving an anesthesiologist in the care of the
ities. The findings are documented, as is the
tumor ablation patient increases the logistics
need for the patient to fast for 6 to 8 hours
of care before, during, and after the procedure,
before the anesthesia (3). Overall, the preanes-
and thereby adds costs to the program. But the
thesia interview addresses the following disease
increase in patient safety and patient satisfac-
areas, which can be included by the ablation
tion, along with the ability to care for patients
team in its routine interview. This would ensure
with more complex conditions, should offset
that patients who are potentially an anesthesia
this cost to the hospital with increased patient
risk would be identified for further consultation
volume and safer practices.
by the anesthesia team.
Once an anesthesiologist is consulted, spe-
cific guidelines toward the care of the patient
Cardiovascular Disease
must be met. These guidelines, codified by the
American Society of Anesthesiology, describe Cardiovascular disease is a risk factor for com-
components for (a) a preoperative visit, (b) plications in patients who are undergoing anes-
physiologic monitoring during the anesthesia, thesia; therefore, any questions that would elicit
(c) a proper postanesthesia care unit for recov- a cardiovascular problem would be appropriate
ery, and (d) a postoperative visit (1). Failure to in the initial history. Although this may include
meet these guidelines would be unsafe for the atherosclerotic stroke, hypertension, or claudi-
patient and would subject the anesthesiology cation, the patients at highest risk for cardio-
practitioner to the risk of malpractice claims. If vascular complications are those who are either
proper planning has not incorporated these in congestive heart failure at the time of proce-
requirements for anesthesia care, the treatment dure or who had a myocardial infarction within
66 lA. Fox and A.M. Harvey

the past 6 months. Accordingly, questions about carbon dioxide production, and potential life-
these conditions need to be asked, and if the threatening arrhythmias. With prompt treat-
anesthesiologist indicates that the patient is at ment with the calcium blocker dantrolene,
high risk for a cardiac event, then one should which should be available wherever anesthesia
consider whether the benefits of the procedure is being administered, malignant hyperthermia
are warranted. Other comorbid conditions that can be stopped. Every institution in which anes-
are markers for cardiovascular disease, such as thesia is given should have a protocol and a kit
diabetes and hypercholesterolemia, also should readily available for the quick treatment of this
be noted (4). rare but potentially life-threatening disorder.

Pulmonary Disease History and Physical Examination of


the Airway
A history of shortness of breath, smoking,
bronchospastic pulmonary disease (asthma) or The anesthesiologist is best qualified to deter-
documented chronic obstructive pulmonary mine by physical examination if a patient has
disease will lead the clinician or anesthesiolo- an abnormal airway that would lead to prob-
gist to document if the patient can withstand lems with ventilation once anesthesia is admin-
positioning for the tumor ablation. This history istered. All anesthesiologists view a history of
would also guide the anesthesiologist in decid- prior airway problems as significant. Specifi-
ing whether other pulmonary function tests and cally, this includes a history of difficult intuba-
other preoperative preparations of the patient tion, a history of mandibular or neck surgery in
are indicated. Patients with an acute upper res- which range of motion of the neck could poten-
piratory infection may be well served by having tially be decreased, or a history of sleep apnea.
their procedure postponed until this acute In the case of sleep apnea, it may be difficult to
event resolves. It is essential to identify ventilate the patient by mask or to secure the
any reversible component of bronchospastic airway by an endotracheal tube once airway
disease so that proper treatment may be initi- reflexes are lost. Here, special precautions to
ated preoperatively and continued intra- and secure the airway in an awake patient may be
postoperatively. needed.
In terms of physical examination, anesthesi-
Anesthesia History ologists look at the length of the mandible
(patients with smaller mandibles are more
Many patients give a history of problems during difficult to intubate), the distance between the
their prior anesthesias. These may include a
mandible and the hyoid bone (smaller distances
history of severe nausea and vomiting postanes- are more difficult to intubate), and a grading of
thesia in which the treatment would include use the view of the hypopharynx when the patient
of antiemetics (ondansetron, ephedrine, meto-
opens the mouth. If there is a large tongue and
clopramide) in the perioperative period or of no view of the soft palate and the epiglottis, this
using of anesthesia agents that are purported
patient is at higher risk for a difficult intubation
to produce less nausea. Some patients give a than are patients with an unencumbered view
history of other severe reactions to anesthesia, of the hypopharynx.
such as cardiac or respiratory arrest. In this case,
The American Society of Anesthesiology has
obtaining the old anesthesia record document- specific recommendations for the potentially
ing this problem would help in avoiding this sit- difficult airway (5).
uation, whatever the cause.
Finally, there is a rare genetic disorder, trig-
gered by many anesthesia drugs, called malig-
Monitoring During the Procedure
nant hyperthermia. This condition, caused by a The American Society of Anesthesiology codi-
disorder of calcium metabolism in the skeletal fied intraoperative monitoring for all anesthe-
muscle, causes a hypercontractile state of the sias about 20 years ago, with a revision in 1998
muscle that results in high fever, increased (6). Monitoring includes a continuous display of
6. Anesthesia for Ablation 67

electrocardiogram (ECG), checking blood the procedure itself, and the amount of postop-
pressure at least every 5 minutes while the erative pain expected. The level of anesthesia is
anesthesia is in progress, and a measure of res- balanced with the stimulus of the procedure.
piratory function. As of 2003, this includes a In analyzing the stimulus for liver tumor
method of capnography and pulse oximetery ablation, there is a body wall component of
for every patient. Finally, an individual trained somatic pain, extending from T5 to T9 skin der-
in the administration of anesthesia medications matomes, where the needles are passed through
should be continuously present throughout the skin, which can be well anesthetized by a
the anesthesia. When asking an anesthesia team long-acting local anesthetic in all patients for
member to be included in the care of the tumor patient comfort. Even if the patient is under
ablation patient, these monitors will need to be general anesthesia, the routine use of a skin
included in the anesthetizing site. local anesthetic is encouraged, as this reduces
the somatic stimuli of the procedure, allows
Postanesthesia Care Unit lower levels of anesthetic agents, and may allow
quicker wake-up. Local anesthetic supplemen-
The postanesthesia care unit, where patients tation may result in less postoperative pain,
recover from their anesthesia, has been shown thus increasing patient satisfaction and facili-
to decrease anesthesia morbidity and mortality. tating discharge.
In this area, personnel who have advanced air- The visceral pain pathways transmit stimuli
way training are available should respiratory from the liver and upper abdominal organs
difficulties arise. Additionally, this area provides during tumor ablation procedures. Visceral pain
a place where additional tests such as postpro- is described as a diffuse, dull, aching, cramping-
cedure chest x-rays, ECG, or blood work can be type pain (7). The celiac plexus innervates most
performed. Discharge from this area should be of the abdominal viscera, including the stomach,
after examination by the anesthesiologist or liver, biliary tract, pancreas, spleen, kidneys,
by the patient meeting protocol criteria for adrenals, omentum, and small and large bowel.
discharge. According to most standard anatomic text-
books, there are three splanchnic nerves-great,
Postoperative Visit lesser, and least (8). The great splanchnic nerve
arises from the roots of T5 or T6 to T9 or T10,
The postoperative visit provides a mechanism
runs paravertebrally into the thorax, through
whereby the patient can give feedback to the
the crus of the diaphragm to enter the abdomi-
anesthesiologist on the conduct of the proce-
nal cavity, and ends in the celiac ganglion on that
dure and anesthesia. During the visit, potential
side. The lesser splanchnic nerve arises from the
problems routinely discussed are the event of
TlO to Tll segments and passes lateral to, or
recall during the procedure, nerve injuries due
with the great nerve to the celiac ganglion. It
to positioning, or other reasons for patient
sends postganglionic fibers to the celiac and
dissatisfaction with the procedure. Pain relief
renal plexuses. The least splanchnic nerve arises
during the procedure and immediately postop-
from Ttl and T12 and passes through the
eration are frequently reviewed for continuous
diaphragm to the celiac ganglion. The plexus
quality improvement.
varies anatomically in relation to the vertebral
column from the bottom ofT12 to the middle of
Pain of the Procedure and the L2. It is important to emphasize that a celiac
plexus block does not provide total anesthesia
Selection of Anesthesia of all upper abdominal visceral sensation or
Techniques reflexes. A cellac plexus block may be combined
with sedation or light general anesthesia, and
The administration of anesthesia involves with local anesthesia/intercostal nerve blocks
assessing the patient's preexisting pain and for the skin for tumor ablation procedures on
anxiety from the medical condition, the pain of the upper abdominal organs.
68 lA. Fox and A.M. Harvey

After needles are inserted, the ablation pro- viscera. It can be accomplished under both
cedure itself produces painful sensations. general anesthesia and local/intravenous anal-
Tumors are ablated by radiofrequency, absolute gesia, progressing to intravenous general anes-
alcohol, and cryotherapy. thesia depending on patient requirements.
Radiofrequency is the heating and destruc- Spinal cord metastases and tumors tend to
tion of tissues in which a high-frequency alter- be treated under local anesthesia and lighter
nating current raises the temperature of the sedation, to allow for patient feedback and
tissues beyond 60°C, resulting in a region of functional assessment of lesion size during
necrosis surrounding the electrode. This ultra- radiofrequency procedures.
sonic energy is an intense stimuli that requires
greater anesthesia levels during the radiofre-
quency procedure. The injection of absolute Types of Anesthesia
alcohol can induce tumor necrosis and shrink-
age by a mechanism of action that involves Anesthesia care for tumor ablation can be
cytoplasmic dehydration with subsequent divided into four types: conscious sedation,
coagulation necrosis and a fiberous reaction. monitored anesthesia care, regional anesthesia,
Absolute alcohol causes a neurologic and and general anesthesia. No hard rules for the
tissue-destructive effect, with significant pain type of anesthesia are presented here. Each
during its administration, thus requiring higher patient is different, and the best anesthesia plan
anesthesia levels. Cryotherapy, which involves is one that is tailored to the procedure and the
the formation of an ice ball and ice crystals with patient's medical condition.
necrosis of tissue, stimulates the pain fibers to
a lesser extent than the other modalities men-
Conscious Sedation
tioned. During the time of administration of
radiofrequency and absolute alcohol, one may Conscious sedation involves the administration
see a rise in pulse rate and blood pressure, indi- of sedative and pain medication by a practi-
cating a sympathetic response to the stimuli. tioner who is not performing the procedure.
Deepening of the anesthesia level or control This practitioner could be a physician or a reg-
of the hypertension and tachycardia by beta istered nurse, and the purpose of conscious
blockade is needed at this point in the anes- sedation is to provide the patient with anxioly-
thesia course. sis as well as pain medication. All conscious
Many centers have been able to accomplish sedation modalities rely on the use of local
ablation of liver tumors under local anesthesia anesthesias by the interventional radiologist
for the skin and body wall and intravenous prior to proceeding. Each institution has
conscious sedation that extends to intravenous established training standards for privileges in
general anesthesia for the visceral pain compo- conscious sedation, which are most likely
nent of the procedure. The requirements of the supervised and written by members of the anes-
procedure, however, suggest that the patient thesiology department. Medications most
should not be moving during the time of needle commonly used for conscious sedation are the
placement to reduce the risk of pneumothorax shorter-acting benzodiazepines (midazolam)
or tearing of the liver capsule during needle and narcotics (fentanyl) because of their rela-
placement, and to provide clearer imaging. tively quick onset and quick offset, along with
General anesthesia, with securing and control their ability to be reversed by the appropriate
of the airway and respiratory movements, may medication (flumazenil and naloxone). Most
offer benefits for these liver tumor ablation institutions set upper limits on the dosage of
cases. conscious sedation medications that can be
Anesthesia for renal tumor ablation is similar given by nonanesthesia staff before a consulta-
to that for liver tumors due to the similar inner- tion by the anesthesiology team is required.
vation of the kidneys and upper abdominal Additionally, if the patients have a history of
6. Anesthesia for Ablation 69

airway complications, a complex medical his- visceral pain pathways. Spinal anesthesia with
tory, or a history of severe anxiety, most hyperbaric local anesthesia to obtain a T4-T5
conscious sedation staff will consult their level also is a possibility. Another regional anes-
anesthesiology backup prior to proceeding. thesia technique is a combination of celiac
plexus block for the visceral component of the
pain with intercostal nerve block for the body
Monitored Anesthesia Care wall and somatic components.
Monitored anesthesia care (MAC) involves a
deeper level of sedation. In this type of care,
more potent hypnotic medications are used
General Anesthesia
alone or in combination, which could com- General anesthesia, during which the patient is
promise the patient's airway or depress the made completely unconscious, requires special-
patient's respiratory drive. Staff who are ized equipment to administer. The practice of
trained in anesthesia care should be present general anesthesia is divided into induction,
whenever a MAC is given, and precautions to maintenance, and emergence. Induction agents
secure the patient's airway should be taken include intravenous drugs like barbiturates
prior to embarking on this type of anesthesia. (thiopental or propofol) or etiomidate. Intra-
Many medications can be used in a MAC from venous induction agents are usually more tol-
increased doses of narcotics (fentanyl) and ben- erated by the patient. There are conditions in
zodiazepenes (midazolam) to the common bar- which an inhalational medication may be
biturate thiopental, or to the newer, shorter indicated to induce general anesthesia. These
acting propofol. inhalational agents, which are halogenated
A common philosophical approach is to hydrocarbons, can be either longer acting or
think in terms of a building block technique in shorter acting, but all are given into the lungs
which a base of anxiolysis and sedation is and taken up into the brain to produce and
achieved with benzodiazepines (midazolam), a maintain unconsciousness. Most of these agents
small amount of narcotic (fentanyl) is given for are monitored by the use of end-tidal expira-
the anticipated discomfort of the positioning tion levels. At the end of the procedure, the
and analgesia for the stimulus of the procedure patient awakens by cessation of continuous
with propofol administration, either as small administration of these medications. Given that
boluses or drug pump infusion technique as the most patients who undergo tumor ablation
fine-tuning to decrease patient awareness. By therapy will need to have multiple CT or MRI
using this building-block technique of short scans, patients who undergo general anesthe-
acting drugs with specific selection of doses sias most likely have their airway secured with
based on the component of anxiolysis, analge- an endotracheal tube, which provides higher
sia, and patient's request for decreased aware- security against aspiration or airway loss while
ness, one is able to more easily maintain the the patient is in the scanner.
patient's spontaneous respiration without res- When indicated, an anesthesiologist may
piratory depression or obstruction. elect to place a device called a laryngeal mask
airway. This tool, which resides in the hypo-
pharynx, can be connected to the anesthesia
Regional Anesthesia machine and, during either spontaneous or
Regional anesthesia can be used for liver tumor controlled ventilation, inhalational anesthesia
ablation procedures once one understands the may be given. Laryngeal mask airways do not
neural pathway of stimulation. Epidural anes- provide total airway protection, but are easily
thesia can be accomplished with a catheter to tolerated by patients. For both endotracheal
control and extend the spread of a local anes- and laryngeal mask airway anesthetics, end-
thesia agent so that T4-Ll is covered. This tidal CO 2 is monitored while the procedure is
would block both the somatic body wall and performed.
70 lA. Fox and A.M. Harvey

Procedure Room Design and labeled outlets connected to an emergency


power supply.
Anesthesia Equipment Selection As a systems philosophy, the anesthesia
and Checkout workstation should be the same configuration
as other anesthetizing locations in the main
In designing facilities for tumor ablation, it is operating room for ease and comfort of use
important to plan for the administration of from a human factor perspective. All anesthe-
anesthesia in a way that meets the American sia locations in the operating rooms are set up
Society of Anesthesiologists' guidelines for with the anesthesia machine placed to the
non-operating-room anesthetizing locations right of the patient's head prior to induction of
(9,10). The location for the anesthesia machine the anesthesia (Fig. 6.1). In the planning of the
should have oxygen and suction outlets as well tumor ablation facilities, consultation with the
as air outlets for ventilator use. A backup anesthetizing team will result in the best work-
supply of oxygen should include the equivalent ing conditions for all involved in patient care.
of at least a full E cylinder. The key monitors in At times, the anesthesiologist may be called
the administration of anesthesia are an ECG, upon to initiate a program within the con-
noninvasive blood pressure (BP) cuff, pulse straints of existing resources. In these situa-
oximeter, and end-tidal CO 2 capnometry. Suffi- tions, where an existing CT scanner room is
cient electrical outlets need to be available for modified as an ablation room (Fig. 6.2), the
these monitors, and there should be a reserve only place for the anesthesia equipment to be
of outlets for other equipment, including clearly placed might be to the left of the patient. If

FIGURE 6.1. An anesthesia workstation in a standard and ambu bag, resides to the right of the patient's
operating suite, illustrating the usual positioning of head. If general anesthesia is induced, a mask fit is
an operating room table in relation to the anesthesi- maintained by the anesthesiologist's left hand while
ologist's workstation. The operating room table is at ventilation is carried on through the ambu with the
the bottom. The anesthesia machine, with its physio- right hand.
logic monitoring screens and attached airway circuit
6. Anesthesia for Ablation 71

FIGURE 6.2. An anesthesia workstation in a com- anesthetizing team. Some anesthesiologists may
puted tomography (CT) scan suite, illustrating a prefer to design an induction room in which general
standard CT scan suite that has been converted into anesthesia is performed and the patient is then
a room for CT-guided ablation procedures. Because moved into the CT scanner, or they may wish to
of space constraints in this conversion, the anesthe- arrange assistance while induction is performed in
sia machine is at the left of the patient's head. This this "reversed" room. With proper communication,
is the reverse from the usual work position of the patients may be anesthetized safely.
anesthesiologist and should be communicated to the

this is necessary, the interventional team must


consult the anesthesia team and inform them of
this situation, as assistance during the induction
of general anesthesia might be required.
Finally, the ideal anesthesia workstation has an
anesthesia cart with drawers for supplies in the
same configuration as any other anesthetizing
location (Fig. 6.3). An MRI-compatible anes-
~
FIGURE 6.3. An anesthesia equipment cart. In addi-
tion to the anesthesia machine, specialized equip-
ment is required that is stored on the cart, as
illustrated here. The top center of the cart usually
holds anesthetic and resuscitative medications that
are drawn into syringes for ready administration. The
other drawers hold airway supplies, intravenous and
central vein placement supplies, and other items
deemed necessary for the conduct of the anesthesia.
Additionally, gloves and a needle disposal box
should be readily available.
72 lA. Fox and A.M. Harvey

thesia machine, MRI-compatible anesthesia is not possible on the radiologic table, then
cart, and equipment should be available for induction and intubation must be accomplished
tumor ablation procedures in the MRI envi- on another stretcher or adjustable surface with
ronment. A large trash disposal for medical the patient then being transferred to the radio-
wastes as well as a wide-mouth, deep, hard-wall logic surface after the airway is in place and
sharps disposal receptacle should be within secured. The patient and monitors need to be
arm's reach for needle safety. visualized during the frequent breath holding
Unique equipment for long-distance admin- and scanning procedures when all personnel
istration of anesthesia includes long breathing leave the room. The ventilator and respiratory
circuit tubing, intravenous tubing extensions, alarms should have both a visual and auditory
end-tidal CO2 sampling tubing extensions, as component so all members of the procedure
well as sufficient lengths of pulse oximetry, and anesthesia teams know when ventilations
noninvasive blood pressure, and ECG cables. have been suspended, either for needle place-
As most anesthestizing locations for tumor ment or radiologic scanning. Adequate illu-
ablation do not have a permanently installed mination of the patient, anesthesia machine,
anesthesia machine, it is even more critical that and monitoring equipment must be available,
the machine and equipment have an appropri- with a backup of a battery-powered form of
ate checkout before each use for key index con- illumination. Distractors in the environment
nection to the medical gas supply (11). The that divert the attention of the anesthesiologist
Food and Drug Administration (FDA) pub- away from the patient's care must be identified.
lished a simplified anesthesia machine checkout Tasks not requiring performance by the anes-
and inspection procedure in 1993 (12). A copy thesiologist should be accomplished by other
of the checkout procedure should be available personnel so that the anesthesiologist can focus
at the point of use, attached to and visible on on the patient and his or her care. Extrane-
the machine. Complete equipment checkout ous noise and tasks need to be reduced to a
and a rigid suction catheter with adequate pres- minimum. For example, door openings and
sure should be present before proceeding closings for the scanning period should be
with patient care. A good guiding philosophy minimized, and table positioning and patient
to prioritize actions for these remote-site positioning tasks need to be unloaded from the
procedures is patient safety is number one, anesthesiologist so that he or she can devote
patient comfort is number two, and on-time full attention to the patient, the monitors, and
performance is number three. An anesthetic airway and anesthesia management.
gas scavenging system should be connected to In establishing the same system of care for
wall suction to meet Occupational Safety and remote-site administration of anesthesia, two-
Health Administration (OSHA) workplace way communication to request assistance with
standards for waste anesthetic gases (13). phone access and phone numbers for anesthe-
Ideally, the waste-scavenging suction should be sia technicians and biomedical support should
a separate outlet and not Y-connected, since the be available at the point of care. As anesthesi-
latter decreases the force of suction power ology is a time-based service and to coordinate
available to clear the patient's airway during procedure events on a timeline, a wall clock
critical airway events. Anesthesia equipment that is visualized by all members of the proce-
should meet current standards. Preventive dure team is important to coordinate timed
and routine maintenance of the anesthesia events. Specialized anesthesia carts for difficult
equipment and monitors with appropriate airway management should be immediately
record keeping should be the same standard available with additional trained personnel to
as for all anesthesia equipment in the assist the anesthesiologists for all off-site anes-
institution. thetizing locations. An emergency cart with
Specific equipment needs include easy access defibrillator, emergency drugs, and other equip-
to the head of the patient for induction and ment adequate to provide cardiopulmonary
intubation, with adjustable table height. If this resuscitation should be available. Code policies
6. Anesthesia for Ablation 73

and procedures with special consideration for a recommended by patient safety advocates and
surgical airway code for the hospital should be required by the Joint Commission on Accredi-
uniform for all off-site locations. Evacuation tation of Realthcare Organizations (JeARO)
procedures should be planned for moving the patient reporting standards.
patient out of the scanner with ambu bag ven-
tilation, oxygen or air ventilation, and portable
physiologic monitoring with pulse oximetry. Complications
Medications to decrease patient awareness,
such as midazolam and propofol, can be admin- The most common complication of these
istered during these unplanned evacuation procedures is pneumothorax. Any inadvertent
procedures. needle insertion into the visceral pleura, during
either liver or renal tumor ablation techniques,
Quality Assurance and can initiate a slow leakage of air from the pul-
monary parenchyma. If the patient is sedated,
Continuous Quality the occurrence may be heralded by the onset
Improvement of chest pain, coughing, shortness of breath, or
pulse oximetry desaturation. With pneumo-
The goals for any quality assurance activity are thorax, physical examination may show either
the improvement in health care and patient hyperresonance or loss of breath sounds on
safety for the patients we serve. For anesthesia the side of the procedure. Agitated, coughing
for tumor ablation, a quality assurance program patients or those with chronic obstructive pul-
should look at the structure, process, function, monary disease (COPD) or emphysema are
and outcomes of patient care (14). Data should at increased risk for pneumothorax. Under
be captured on all adverse events and near-miss general anesthesia, suspending positive pres-
events so that an interdisciplinary team of inter- sure ventilation so the lungs are deflated at the
ventional radiologists, anesthesiologists, nurses, time of needle and radiofrequency probe or
and administrators can analyze and improve cryoprobe placement may decrease the risk of
the tumor ablation system of care. After any this event. If a pneumothorax is suspected,
adverse event, patient data are retrieved, and positive pressure ventilation and nitrous oxide
the members of the tumor ablation team are may worsen the pneumothorax and cause the
interviewed and debriefed to obtain the various development of a tension pneumothorax. If
perspectives of the case and event. A timeline the needle passage is close to the diaphragm or
is created so that all observations and multiple signs and symptoms of pneumothorax occur, as
dimensions of contributing factors can be the patient is already in the scanning radiologic
placed in this two-dimensional structure. A device, imaging to rule out a pneumothorax is
physiologic theory of the event or injury is warranted. With lung tumor ablation, pneu-
created, along with a systems theory that ana- mothorax is anticipated.
lyzes the contribution of the environment of A second potential complication is position-
care and human factors to the event. Anytime ing injuries to nerves (16,17). Care should be
equipment malfunction is suspected in adverse taken whenever the angulation of the arms and
patient events in remote locations, an impartial shoulders is greater than 90 degrees axially,
investigation protocol should be followed for as this puts stretch forces on the brachial
analysis of the event (15). The literature is plexus. Anesthetized patients are susceptible
reviewed to fully define the event and see if this to brachial plexus injury as they are unable to
event has been described before. The team then respond to pain, numbness, paresthesia, or
makes its recommendations for improved care weakness. Muscle relaxants used during these
in a nonpunitive educational approach, with the procedures may reduce the protective effect of
systems of care improved so that the event does normal muscle tone against nerve stretch
not recur. The patient and family are fully injuries. Strategies to prevent brachial plexus
informed of medical events, as is currently injuries include positioning the arms across the
74 lA. Fox and A.M. Harvey

chest with adequate padding to slightly raise 20999, lung CPT 32999, bone CPT 20999,
them from skin contact of the chest wall, posi- soft tissue CPT 20999, and spinal cord CPT 99)
tioning the arms above the head only during the have to use a nonspecific "99" code with
actual imaging stage, and frequently chang- a specific detailed report sent with the submit-
ing the arm position, especially in prolonged ted encounter form and bill (18). The guidance
procedures. codes that also are submitted include a CT
guidance code (CPT 76360), MRI guidance
(CPT 76393), and ultrasound guidance (CPT
Reimbursement for Providing 76942).
The associated common CPT codes for
Anesthesia Services anesthesia for liver tumor ablation is 00790,
with a seven-base-units-plus-time billing. The
Adequate reimbursement for anesthesia ser- American Society of Anesthesiologists (ASA)
vices needs to be considered in order to obtain also has a relative value guide (RVG), which is
the resources of an anesthesiologist in the care also used for billing (19). The ASA RVG
of these patients. One needs to understand code for anesthesia for intraperitoneal upper
anesthesia billing for tumor ablation services so abdominal procedures is 00790, also of seven-
that the patient, procedure, and time of the case base-unit-plus-time valuation. Anesthesia for
can be documented and income can be gener- tumor ablation for renal lesions is ASA code
ated. Anesthesia is a time-based service as the 00862; anesthesia for tumor ablation for a lung
anesthesiologist has no control over the length lesion is ASA code 00520; and for anesthesia
of the procedure. Typically, the procedure has a for tumor ablation for spinal cord lesions 99;
"base" unit value, which includes the value of the cervical ASA code is 00600, the thoracic
all usual anesthesia services except the time ASA code is 00620, and the lumbar ASA code
actually spent in delivering anesthesia care. The is 00630.
base units include the usual preoperative and In the submission of anesthesia billing, it
postoperative visits, the preoperative introduc- is important to accurately define the anesthe-
tion, review of chart, IV start, administration of sia services provided. General anesthesia is
fluids incident to the anesthesia care, and inter- defined as a loss of consciousness and protec-
pretation of monitoring (ECG, BP, oximetry, tive reflexes; MAC is an intermediate plane of
capnography, gas monitoring, and tempera- anesthesia; and conscious sedation is a lighter
ture). The "start time" and time units begin plane of anesthesia, in which patients can
when the anesthesiologist is in constant per- respond to verbal commands and can protect
sonal attendance, and time is measured in 15- their own airway. Conscious sedation provided
minute intervals. One time unit is 15 minutes or by nonanesthesiologists, such as an indepen-
a portion thereof. The "end time" is when the dent trained person to assist the physician in
patient is signed out to the postanesthesia care monitoring the patient's level of consciousness
unit (PACU), with intensive care unit (ICU) and physiologic status, can be billed under CPT
nursing care or its equivalent. codes 99141 (intravenous, intramuscular, or
Facility and interventional radiologists will inhalational) and CPT 99142 (oral, rectal, or
typically use two codes for submission, a surgi- intranasal).
calor procedure component code and the guid- Medicare and some private payers need
ance code. Only two tumor ablation codes are to have justification for MAC billing with
specific at this time for liver tumor ablation, regard to medical status, ASA classification,
that being for Current Procedural Terminology underlying medical disease, anxiety, or inability
(CPT) code 47382, ablation, one or more to accomplish the procedure under conscious
tumor(s), percutaneous, radiofrequency; and sedation analgesia before the payer will
CPT 47381, ablation, one or more tumor(s), reimburse for MAC services. Constant moni-
cryosurgical. All other sites for tumor ablation toring of the billing and receipts for anesthetic
(abdomen CPT 20999, retroperitoneal CPT procedures for tumor ablation are needed to
6. Anesthesia for Ablation 75

assure clinic income to support the anesthesia 10. Dorsch JA, Dorsch SE. Operating room design
needs of the patient undergoing these proce- and equipment selection. In: Understanding
dures and to maintain the services of an anes- Anesthesia Equipment. 4th ed. Baltimore:
thesiologist to assist in anesthesia for tumor Williams & Wilkins, 1999:1015-1036.
ablation. 11. Dorsch JA, Dorsch SE. Equipment checking
and maintenance. In: Understanding Anesthesia
Equipment. 4th ed. Baltimore: Williams &
References Wilkins, 1999:937-965.
12. Morrison JL. FDA anesthesia apparatus check-
1. Standards of the American Society of Anesthe- out recommendations, 1993. American Society of
siologists: basic standards for preanesthetic Anesthesiologists Newletter 1994;58(6): 25-26.
care (1987), standards for post anesthesia care 13. U.S. Department of Health, Education, and
(1994). ASA Executive Office, 520 N. Northwest Welfare. Criteria for a recommended standard;
Highway, Park Ridge, IL 60068-2573. occupational exposure to waste anesthesia
2. Practice advisory for preanesthetic evaluation. gases and vapors. (DHEW [NIOSH] publication
Anesthesiology 2002;96:485-496. No.77-140). Washington, DC: US Government
3. Warner MA, et a1. Practice guidelines for preop- Printing Office, 1977.
erative fasting. Park Ridge, IL: American Society 14. Dodson BA. In: Miller RA, ed. Quality Assur-
of Anesthesiologists. Anesthesiology 1999;90: ance/Quality Improvement in Anesthesia. 5th
896-905. ed. New York: Churchill Livingstone, 2000:
4. Roizen MA, Foss JF, Fischer SP. Peroperative 2597-2612.
evaluation. In: Miller RA, ed. Anesthesia. 5th ed. 15. Dorsch JA, Dorsch SE. Chapter 26. In: Under-
New York: Churchill Livingstone, 2000:824-883. standing Anesthesia Equipment. 4th ed. Balti-
5. Practice guidelines for management of the more: Williams & Wilkins, 1999:960-961.
difficult airway. Park Ridge, IL: American 16. Warner MA, et a1. Practice advisory for the
Society of Anesthesiologists. Anesthesiology prevention of perioperative peripheral neu-
2003. www.asahq.org ropathies. Park Ridge, IL: American Society of
6. Standards for basic anesthesia monitoring. Anesthesiologists. Anesthesiology 2000;92:1168-
1998. ASA Executive Office, 520 N. Northwest 1182.
Highway, Park Ridge, IL 60068-2573. 17. Shankar S, vanSonnenberg E, Silverman SG,
7. Bonica JJ. In: Loeser JD, ed. Management Tuncali K, Flanagan HL, Whang EE. Brachial
of Pain. Philadelphia: Lippincott Williams & plexus injury from CT-guided RF ablation under
Wilkins, 2001;679:1293-1296. general anesthesia. (CVIR in press)
8. Cousins MJ, Bridenbaugh Po. Neural Blockade 18. CPT 2003. American Medical Association, 515 N.
in Clinical Anesthesia and Management of Pain. State Street, Chicago IL 60610.
3rd ed. Philadelphia: Lippincott-Raven, 1998: 19. ASA Relative Value Guide, 2003. ASA Execu-
463-470. tive Office, 520 N. Northwest Highway, Park
9. Guidelines for nonoperating room anesthetizing Ridge, IL 60068-2573.
locations. 1994. ASA Executive Office, 520 N.
Northwest Highway, Park Ridge, IL 60068-2573.
7
Devices, Equipment, and Operation of
Ablation Systems
Paul R. Morrison and Eric vanSonnenberg

This chapter reviews the operation of each of time. Importantly, such data affect the course of
several medical systems that is used to deliver the procedure, as they are indicative of whether
local thermal therapy. Each system provides or not a therapeutic end point has been
its own mechanism for the transfer of energy reached; for example, a planned protocol might
within tissue that results in cytotoxic effects. be altered should the temperature reading be
This targeted destruction of cells is referred to too low, the resistance too high, or the time too
as tissue ablation. This chapter concentrates on short.
the medical system itself, and not the principles
behind the various mechanisms of tissue abla-
tion. It is concerned with the physician as a user,
or operator, of the ablation system during a pro- Radiofrequency Ablation
cedure. Accordingly, emphasis is placed on the Systems
following: the thermal applicator, the main
control panel, user-defined parameters and set- Radiofrequency ablation (RFA) is performed
tings, and feedback to the operator. with the patient and an RF generator serving as
The applicator is the "working end" of the the primary components of an electrical circuit.
system. Examples include an electrode wired The generator is the source of current in the
to a radiofrequency (RF) generator, a light- circuit and the human body serves as a resistor,
diffusing tip on an optical fiber connected to a all connected by wire leads. It is the resistance
infrared laser, a microwave antenna, a cryo- in the tissue to the RF current that generates
genic probe, and a high-intensity ultrasound heat and creates a thermal ablation effect. The
transducer. The "thinking end" of the system electrical resistance of the body's tissue is
includes a control panel, by which the user can referred to as the impedance, and is measured
communicate with the device to conduct the in ohms (0). Impedance, rather than resistance,
ablation. The control panel is used by the oper- is a term more appropriate when dealing with
ator to set and adjust treatment parameters that alternating current.
include the energy delivered, the rate of energy In monopolar-type RFA systems, the abla-
deposition, the duration of an exposure, or the tion is confined to a targeted volume of tissue
temperatures within tissue. Observation of by inserting an RF electrode that is connected
these and other parameters on the control to the generator directly into the tissue. The
panel during ablation serves as data feedback electrically noninsulated tip of the electrode is
to the user on the progress of the procedure. relatively small, and thus the current density
This feedback may be an electronic display of surrounding this tip is high; this creates a local-
the temperature of the applicator, the electrical ized thermal effect due to ionic agitation. A
resistance of the tissue, or simply the elapsed return connection to the RF generator is

76
7. Devices, Equipment, and Operation of Ablation Systems 77

required to complete the circuit. This return is The RFA systems produced by several man-
established by placing dispersive electrodes in ufacturers are presented in the following sec-
contact with the skin, usually on the patient's tions. Manufacturers are presented not in
thighs. These grounding pads offer a large alphabetical order, but rather in chronologic
surface area, keeping the current density low to order based on when the technology first
eliminate heating at the sites of contact. became available in the marketplace.
For tumor ablation, generally, there are two
types of monopolar RFA electrodes: needle and
array. As noted above, both provide segments
Radionics
of a noninsulated conductor that can be placed The Radionics RFA electrode (Cool-Tip,
into tissue and through which the RF current Radionics, Burlington, MA) is shown in Figure
flows. The needle-type electrode is a thin cylin- 7.1. The single 17-gauge electrode is a needle-
drical conductor, usually beveled at the tip, type design. The overall length of the electrode
making it sharp so as to penetrate into tissue. It is selected (10, 15,20, or 25 cm) to provide the
is electrically insulated along its shaft up to the appropriate depth of penetration into the body.
exposed tip; the latter is usually 1cm or a few Separate consideration is given to the selection
centimeters long, which is referred to as the of the active length of the electrode (1, 2, or
"active length". This tip provides an extended 3 cm). As noted above, this choice of the length
source of heat (compared to a single point of the exposed tip establishes the physical
source just at the very tip), the length of which dimension of the RF source, allowing the user
can be selected to best match the dimensions of to best match the ablation to the target. The
the target. The array-type electrode also serves Radionics cluster electrode, also shown in
as an extended source; from a needle-like Figure 7.1, consists of three single electrodes
cannula a number of individual noninsulated fixed in one handle, each with a 2.5-cm exposed
wire electrodes splay out radially into tissue tip. The cluster design adds to the extended
away from the long axis of the cannula. The source of RF energy and is intended to yield a
diameter of the radial extension also can be volume of ablation (on the order of 4 cm in
chosen to best match the target volume. diameter), larger than that for a single elec-

FIGURE 7.1. Valleylab needle-type RF electrodes. A The "cluster" electrode (bottom) combines three
"single" electrode (top) with a 3-cm noninsulated single electrodes with 2.5-cm active lengths to induce
active length is internally cooled during ablation by larger volume lesions. Needle-like tips provide pen-
a closed-loop flow of water. Cooling reduces tissue etration into tissue (inset). (Photos printed with per-
char that might impede RF current flow into tissue. mission from Valleylab.)
78 P.R. Morrison and E. vanSonnenberg

A B
FiGURE 7.2. Valleylab RF generator and coolant Wires lead from panel to electrode and grounding
pump. Running in "impedance control" mode, power pads. A rotary pump (B) sends water to the elec-
is adjusted automatically by the system to maintain trode. Water-cooled tips read cool water tempera-
the level of baseline tissue impedance measured in tures, 22°C in the figure. The ablation end point is
ohms. The main control panel (A) displays imped- based on time, that is, duration of exposure. (Photos
ance, elapsed time, current, power, and temperature. printed with permission from Valleylab.)

trode. The device operates at a frequency of 480 in this mode. The power and current are auto-
kHz (1). matically lowered to reverse any large increase
A salient feature of the Radionics Cool-Tip in impedance. With this device, the latter is
system is that, as the name suggests, the elec- understood to indicate gas formation in tissue
trode is cooled by a continuous flow of cold that is counterproductive to the conduction of
water (-20 De). This flow is done in a closed RF into tissue.
loop; no water is deposited into tissue as the Consider these sample readings from the
water flows internally to, and returns from, the main control panel in Figure 7.2 at 3 minutes of
tip. The cool tip temperatures are intended to RFA: 720, 1.49 A, 143W, 22 DC. Note that the
prevent excessive heating and carbonization of temperature reading is low due to the cool
tissue immediately adjacent to the electrode water flowing through the probe. Thus, while
that can pose high impedance to current flow the tissue around the probe is heated, there is
and prevent optimal penetration of the energy. no temperature feedback that provides a
The generator and coolant pump are shown measure of the elevated temperatures. The end
in Figure 7.2. The main control panel on the point for the procedure is, as noted above,
generator includes visual displays of the tissue a duration (i.e., 12 minutes). The tumor tissue is
impedance (0), the current (amps), the power indeed heated, as there is considerable energy
(watts, W), probe temperature (I), and the being deposited. The heating is observed at the
elapsed time (minutes). There is a potentiome- end of the procedure just prior to the end of the
ter (knob) to control the power output. exposure when the coolant flow is turned off to
The device has a switch on the main control expose tissues immediately adjacent to the
panel that allows the user to select a mode of electrode to an uncooled probe. With the pump
operation. The device typically is operated in off, the static water in the electrode quickly
impedance control mode. The coolant pump is comes to equilibrium with the tissue, and tem-
switched on, a timer on the main control panel peratures on the order of lOODC typically are
is started, and the power knob is rotated to its seen.
maximum. The ablation at that location is com- Alternatively, the device can be operated in
pleted when the timer reaches a set time (12 a manual mode to control the output via manip-
minutes). There are no adjustments made by ulation of the power knob. No coolant is used
the operator to any parameters during ablation and the electrode's temperature is read on the
7. Devices, Equipment, and Operation of Ablation Systems 79

main control panel. This manual mode typically


is used for the brief coagulation of the access
tract as the electrode is withdrawn. The power
is increased until the temperature reading indi-
cates immediate coagulation (usually targeting
near or above 80°C), and then the electrode is
pulled back through the normal tissues. Before
complete removal from the body, the power is
turned off. In addition to tract coagulation,
manual mode without cooling can be used to
perform RFA of tumors in which both time and
temperature are provided as feedback to the
operator.
FIGURE 7.4. Boston Scientific Corporation-Oncol-
Boston Scientific ogy RF generator. The main control panel displays
the elapsed time, power, and tissue impedance. With
The LeVeen needle electrode (Boston Scien- this device, the power is manually increased stepwise
tific Corporation-Oncology, Natick, MA) is an over time. The end point for tissue ablation is a
array-type electrode. It consists of a 14-gauge marked rise in impedance above baseline, e.g., to 467
bevel-tipped cannula (12, 15, or 25cm long) ohms from a baseline of 50 at 12 minutes as seen in
fixed to a handle. Within the cannula are the the figure. The power automatically ramps down
individual elements of the array that are when the end point is achieved. (Photo printed with
deployed from the tip into tissue by a driving permission from Boston Scientific Corporation.)
mechanism in the handle. The electrode is
shown in Figure 7.3. On deployment, the tines arate noninsulated element of the electrode.
of the array splay radially out and arch back, The array diameters can be selected (2, 3, 3.5, 4
each in a semicircular fashion, forming an or 5 cm) to match the tumor volume. The elec-
umbrella-like pattern. Each tine serves as a sep- trode is powered and controlled by the RF 3000
generator shown in Figure 7.4 that operates
at a frequency of 460kHz and provides up to
200 W of power (2). As with most of the RF sys-
tems under discussion, the electrical circuit is
completed with the attachment of four return
electrodes (grounding pads) to the patient.
The main control panel of the generator has
primary displays for time, power, and imped-
ance. There are also control keypads for adjust-
ing the power, setting a time duration, starting,
stopping, and resetting the system. On power-
ing up the generator, there is a series of quick
self-checks leaving it in a ready mode. When the
start button is pressed, it enters into an operat-
ing mode. A small blue light on the panel and
an audible tone serve as indicators that RF
FIGURE 7.3. Boston Scientific Corporation-Oncol-
ogy array-type RF electrode. The deployed array of
energy is being delivered. The operator sets
multiple noninsulated tines establishes an extended the power to a starting value (e.g., 50W for a
source of RF in tissue, here 4cm in diameter. The 3.5-cm array). Per the manufacturer's operating
electrode is initially placed into tissue with the tines instructions this is increased manually over
withdrawn into the cannula. (Photo printed with per- time to a preestablished maximum value (i.e.,
mission from Boston Scientific Corporation.) by lOW per minute to a 100 W limit).
80 P.R. Morrison and E. vanSonnenberg

The signature feature of the system is its allows temperatures at multiple sites of the
impedance feedback mechanism. The elec- array to be monitored, and (c) the XLi version
trode's geometry and the incremental power- provides for the infusion of hypertonic saline
adjustment algorithm allow tissue impedance through select elements of the array. The device
to serve as an end point for the ablation. With operates at 460kHz and up to 100W (3).
this device, the rise in impedance is due to The StarBurst XL electrode is composed of
tissue coagulation and desiccation around the a 14-gauge trocar (10, 15,25 cm long) attached
array. No measure of temperature is provided to a handle. The device is introduced into the
by this device. While the time is marked by patient with the tines of the array retracted
the user, it serves as a guide for each power within the cannula. When properly situated in
increase and does not dictate the duration of tissue, a mechanism on the user end of the
the ablation. Overall, the impedance changes handle deploys the tines. The nine tines splay
little, if at all, from its initial value when the forward and radially out from the tip. The array
electrode is activated. Generally, after many is intended to provide a 3- to 5-cm-diameter
minutes, the impedance rises substantially and volume of ablation; a smaller IS-gauge seven-
rapidly (i.e., by tens and then by hundreds of tine version provides 2- to 3-cm-diameter abla-
ohms in less than a minute). This is known casu- tions. Each array includes tines containing
ally as "roll-off", and the power automatically thermocouples by which tip temperatures are
drops to near zero. Figure 7.4 shows the system continuously measured. The nine-tine array has
at the end of an ablation. Notably, the manu- five thermocouples.
facturer's instructions call for a second phase The XLi version of the electrode provides
of treatment after roll-off ("booster"). Without saline infusion into the tissue during RF abla-
a change in electrode location, the generator is tion. The XLi electrode is shown in Figure 7.5.
activated again at 70% power, until a repeat This feature is intended to provide for larger
roll-off is achieved. ablations (up to 7 cm) using the introduction of
saline in the tissue as an extended source for
the RF current deposition due to its conductiv-
RITA ity. The slow flow of saline is controlled by a
The RFA system of RITA Medical Systems, separate syringe pump and control unit.
Inc. (Mountain View, CA) is composed of the The main control panel of the RITA model
model 1500X RF generator and the StarBurst 1500X includes readouts and controls for tem-
XL and XLi electrodes. The salient features of perature, power, and impedance, as shown in
the system are: (a) the applicator is an array- Figure 7.6. Prominently featured on the panel
type electrode, (b) one mode of operation are the displays for the probes' thermocouples

B
FIGURE 7.5. RITA Medical Systems, Inc. array-type the tissue from the central tine (B) to provide an
RF electrode. These array-type electrodes (A) extended RF source for larger ablation volumes.
include thermocouples in a number of the tines The Xli Electrode shown is intended to create 7cm
by which separate temperature measurements are diameter lesions. (Photos courtesy of RITA Medical
obtained for ablation control. Also, in the XLi Systems, Inc.)
version of the device, saline can be infused into
7. Devices. Equipment, and Operation of Ablation Systems 81

Alternatively, the system can be run in a


mode that allows the user to manually control
the power output to the electrode. This can
serve as a technique for volume ablation of
tissue, or this can provide for coagulation along
the electrode's tract as it is withdrawn from
tissue. The latter is accomplished by heat emis-
sion from a noninsulated portion of the
cannula's tip with the tines undeployed. The
company recently developed a flexible trocar
shaft for improved entry and monitoring for
CT-guided ablations.

FIGURE 7.6. RITA Medical Systems, Inc. RF genera-


tor. The main control panel displays user-defined set- Berchtold
tings for the target tissue temperature, power, and The Berchtold HiTT electrode (Berchtold,
time. Feedback to the user includes the actual RF
GmbH, Tuttlingen, Germany) is a needle-type
power being delivered, the elapsed time, and tissue
impedance. Up to eight thermocouple measurements electrode designed to provide saline infusion
to assess the temperature at the tines of the array (up (4). That company identifies this method
to four) and at separate locations targeted with as high-frequency induced thermo-treatment
added needle-thermocouples can be displayed. In (HiTT). Saline infusion is used to provide addi-
the figure, as an example, the device controls power tional thermal and electrical conductivity in the
to give average temperature readings of 70°C in four tissue to enhance the thermal effect, that is, to
tines of an array. Temperature and time serve as the provide large ablation volumes up to 5 cm in
end points for ablation. (Photo courtesy of RITA diameter. The saline is not intended to provide
Medical Systems, Inc.) cooling for the electrode. A series of microholes
have been drilled in the tip of the probe to
by which up to five temperatures can be moni- deliver the saline. The HiTT electrodes are
tored. A signature mode of operation for the manufactured with active lengths of 1, 1.5, and
device is its automatic temperature control 2 cm. These are 1.2 to 2.0 mm in diameter, and
mode. In this mode, a target temperature is range from 10 to 20cm long. A sample elec-
chosen. The electrode's power is controlled by trode is shown in Figure 7.7.
an internal computer algorithm whereby it The Elektrotom HiTT 106 generator oper-
increases the power until the thermocouples' ates at a frequency of 375 kHz and provides
readings average the set temperature (i.e., power up to 60 W. It is shown in Figure 7.8 along
90°C). The power is then adjusted automati- with the fluid pump used to control the flow
cally to maintain t):lis temperature. A timer of saline. The generator's main control panel
counts from the time this temperature is displays the temperature, flow rate (mL/hr),
reached. The end point for the ablation is a 10- power, energy, impedance, and elapsed time.
minute duration at the set temperature. Addi- For automatic control of the power output, the
tional temperature information can be gained device can be run in either impedance control
by using add-on, nonarray needle thermocou- or temperature control mode. The temperature
ples that can be monitored; there are three control mode can operate on readings from the
extra readouts on the main control panel for temperature sensor in the electrode, or from a
this purpose. In addition to a temperature separate temperature sensor that can be placed
control based on the average of temperature into the tissue for a reading remote from the
values observed, the device can operate in a electrode. A separate manual mode provides
highest or lowest set-temperature mode to the operator with control over the power for
keep temperatures below or above some pre- coagulation of the electrode's tract on removal
scribed value, respectively. from the site of ablation. The infusion pump can
82 P.R. Morrison and E. vanSonnenberg

A B
FIGURE 7.7. Berchtold GmbH & Co. needle-type provide an extended RF source for enhancement
electrode (A). Microholes along the tip of the of the RF ablation. (Photos courtesy of Berchtold
electrode (B) allow saline infusion into tissue to GmbH & Co.)

be controlled manually, as well as automati- needle-type RF electrodes. These differ from


cally. In automatic mode, the flow depends on the above-mentioned RFA devices in that they
the power setting of the unit at the outset of are bipolar electrodes (5). In the bipolar con-
ablation. figuration, instead of an electrical circuit estab-
lished from the needle-electrode through tissue
to the grounding pads, both electrode elements
Celon are situated at the needle tip. RF current flows
The Celon ProSurge applicators (Celon AG between these elements and thus only in the
Medical Instruments, Berlin, Germany) are vicinity of the device's tip. No grounding pads
are used with this device. Celon electrodes are
available in a range of styles for a variety of
ablation procedures. They have active lengths
ranging from 9mm to 3 cm, shaft lengths from
10 to 120 cm (the latter for endoscopic use), and
diameters range from 1.3 to 1.8mm. The Pro-
Surge line of electrodes for interstitial volume
ablation is internally cooled (used in conjunc-
tion with a coolant pump), and intended to
provide ablations 2cm wide with the length
depending on the choice of active length
(1.7-3.0cm) of the device. These are 1.8mm in
diameter, and 10, 15, or 20cm long. A sample
Celon ProSurge electrode is shown in Figure 7.9.
The CelonLab PRECISION generator is
shown in Figure 7.10. The unit operates at 470
kHz. The power available from the unit (range
1-25 W) is relatively low due to the efficiency
FIGURE 7.8. Berchtold GmbH & Co. RF generator of the bipolar system. The main control panel
and infusion pump. Saline is delivered to the RF features readouts of power, time, and imped-
electrode for infusion into tissue by the pump sitting ance. The system runs in an impedance control
atop the RF generator (A). The main control panel
mode. An initial power is set, impedance is
of the generator displays temperature, flow rate of
the saline, power, and time (B). The system can be
monitored, and an acoustic display provides
operated in either an impedance control or temper- an audible tone at a frequency that increases in
ature control mode, whereby feedback to the user proportion to tissue impedance. The ablation
would be impedance in the tissue, or time and tem- end point is reached when the tone changes to
perature, respectively. (Photos courtesy of Berchtold a pulsed tone, indicating that the optimum
GmbH & Co.) impedance has been reached.
7. Devices, Equipment, and Operation of Ablation Systems 83

A B
FIGURE 7.9. Celon AG Medical Instruments needle- tissue coagulation. No grounding pads are connected
type RF electrode. This bipolar device (A) includes to the patient. An internal, closed-loop flow of water
both positive and negative electrode elements at the can be used to cool the tip to reduce tissue char that
tip (B). Electrical current flows between these ele- can impede RF current flow. (Photos printed with
ments and generates resistive heating that results in permission from Celon AG Medical Instruments.)

Cryoablation Systems designed to penetrate tissue. Each cryoprobe is


incorporated into a handle that is in turn teth-
Medical systems designed for cryoablation of ered to the main component of the system for
tumors utilize applicators of various geometries the cryogenic medium to pass into the probe.
that are placed in contact with tissue to subject Contemporary systems freeze tissue using
cells to freezing at lethal temperatures. For an argon gas-based cryogenic technique to
interstitial therapy, the probes are needle-like, achieve low temperatures. High-pressure argon
gas is driven through a very thin tube situated
inside the probe, and oriented coaxially to the
probe's outer hull. Gas exits through a small
aperture at the end of the tube near the probe's
tip. The argon gas returns to the system in a
closed loop. No gas is deposited into the tissue.
Thus, the argon gas undergoes a change from
a high pressure of approximately 3000 psi
(pounds per square inch) to a relatively low 200
psi. In accordance with the Joule-Thompson
effect, this results in a decrease in argon's tem-
perature to freezing values (as low as -180°C).
The use of a gas as the cryogen enables the
design and manufacture of relatively small
probes and allows for the use and control of
multiple probes simultaneously.
FIGURE 7.10. Celon AG Medical Instruments RF
These gas-based systems take advantage of
generator. The main control panel displays the power
output by the device and the elapsed time. It oper- the markedly different behavior of gaseous
ates in an impedance control mode. An audible tone helium. Under essentially the same conditions
provides the user with an indication that the tissue as for argon above, due to its different physical
impedance has markedly increased, indicating com- properties, high-pressure helium undergoes an
plete ablation around the tip. (Photo printed with increase in temperature on passing through the
permission from Celon AG Medical Instruments.) aperture. Thus, while passing argon through the
84 p.R. Morrison and E. vanSonnenberg

probe provides freezing temperatures, passing


helium enables the probe to be heated. This
allows for quick freezing and thawing by the
argon and helium, respectively. This can be used
for immediate control of the freezing process,
as well as for the quick release of the probe
from frozen tissue. The latter assists in the repo-
sitioning of probes to treat multiple sites during
a procedure.

Galil
The CryoHit ablation system (Galil Medical,
Westbury, NY) is gas-based and uses argon and
helium as described above (6). While cry-
oprobes are available for direct surface contact
(having rounded hemispherical tips or flat
ends), a needle-type design also is available for
interstitial use. The latter probes are sized at
1.5, 2.4, and 3.2mm in diameter with lengths
ranging from 17 to 40 cm. A probe is shown in
Figure 7.11. The CryoHit system is shown in
Figure 7.12. The system is not a tabletop device,
but rather is a free-standing wheeled unit.
Notably, it must be located so as to be able to
FIGURE 7.11. Galil Medical needle-type cryoablation
connect to tanks that provide gases during the
probe. The figure shows a 2.4-mm-diameter probe
procedure. Up to seven probes can be used
designed for percutaneous tumor ablation. High-
simultaneously with the system. pressure argon gas flows in a closed loop to the probe
The main control panel is accessed through tip (inset) where it undergoes a thermodynamic
a standard computer monitor with operations process and generates freezing temperatures in
controlled by a keyboard and touch-pad mouse. tissue. Alternatively, helium gas can be used,
The software provides a menu of screens for the whereby the tissue can be warmed and thawed.
user to enter information or observe treatment
parameters. Included are screens for preproce-
dural entries, one of which allows for clinical Figure 7.12. It shows the temperature readings
data such as the physician's name, patient of the probes during the freezing and thawing
name, hospital record number, sex, date of processes and the elapsed times. Probes can be
birth, and history to be recorded. The Targets individually controlled via this screen and set to
Setup screen includes choices for the target stick, freeze, thaw, or off (inactive, no gas flow).
temperatures for all primary modes of opera- Target values can be modified during ablation
tion for each cryoprobe: freeze, thaw, and stick. to control individual probes. Temperature and
Typical choices are -lS0°C for freezing and time provide the primary feedback to the user.
+35°C for thawing. The additional stick mode Treatments are composed of a timed freeze
(typically -20°e) is used to provide a limited (usually 15 minutes) with a thaw; some practi-
freeze to the probe, whereby the probe attaches tioners use a repeat freeze after the thaw.
to the immediate surrounding tissue, and pre-
vents migration of the probe while other probes
are placed.
Endocare
The primary interface during the procedure The CRYOcare system (Endocare, Inc., Irvine,
is the General Operations screen shown in CA) for cryoablation is also an argon/helium-
7. Devices, Equipment, and Operation of Ablation Systems 85

A B
FIGURE 7.12. Galil Medical cryoablation system. The controlled independently. The display also indicates
system (A) requires connections to tanks and reser- the status of each probe (stick, freeze, thaw, off).
voirs of argon and helium gases. For each of up Here, a five-probe freeze is ongoing and viewed after
to seven probes, the main control console (B) dis- approximately 13 minutes with a temperature
plays the selected target temperature, the actual average of -128°C.
temperature, and the elapsed time. Each can be

based system. Probes are available with diame-


ters of 2 to 8 mm, a range intended to cover
both percutaneous and open surgical pro-
cedures (i.e., prostate and liver ablation,
respectively). An example of these needle-type
probes is shown in Figure 7.13. The four-probe
cryoablation system is shown in Figure 7.14; an
eight-probe system also is available.
The main control panel is displayed on a com-
puter monitor. Multiple probes can be operated
simultaneously with individual control. The
status of each probe is displayed, and indicates
whether the probe is in stick, freeze, thaw, or off
mode. The panel displays each tip's temperature
and elapsed time at a given status. Duration and
depth of the freeze serve as feedback to the FIGURE 7.13. Endocare, Inc. needle-type cryoabla-
tion probe. Argon and helium gases are used to
physician. The default setting for stick mode is
provide freezing and thawing of the ablation probe.
-lOoC, and thaw is +40°C; typical temperatures
Utilizing gases for temperature control allows for
recorded during the freeze cycle are between quick reversal of freezing to control ablation, and for
-1300 and -IS0°C. release of the probe from frozen tissue for reposi-
The freeze temperature can be adjusted by tioning. A sample in vitro freeze demonstrates the
modifying the duty cycle of the argon gas, refer- ice ball formed at the probe tip (inset). (Photos cour-
ring to the time over which gas is sent to the tesy of Endocare, Inc.)
86 P.R. Morrison and E. vanSonnenberg

A B
FIGURE 7.14. Endocare, Inc. cryoablation system. The tip temperature, and the elapsed time. In the figure,
system (A) supports the use of multiple probes simul- probes are disconnected and read as "open", whereas
taneously. For each probe, the main control console connected probes would be active and display values.
(B) displays the probe's status (off, stick, freeze, Temperature and time are selected to effect an abla-
thaw), the flow duty cycle, target temperature, actual tion. (Photos courtesy of Endocare, Inc.)

probe. A duty cycle of 50% would adjust the contact applicators. Contact tips touch tissue
flow of gas to be on-then-off half the time, and are used for thermal resection (laser
respectively, allowing the physician to control scalpel) or interstitial coagulation in situ. Inter-
the rate of ice formation. In addition to the tem- stitial laser ablation can be done with a simple
peratures of each of the cryoprobes, the user bare fiber optic inserted into the tissue. A more
can add and monitor separate individual needle contemporary interstitial approach uses an
thermocouples for additional temperature extended light source, the diffusing tip. It acts
measurements at sites remote from the probes as a cylindrical light source whereby light is
during the procedure. emitted radially along the end of the tip to
provide a volume of ablation larger than that of
the simple bare fiber. These fibers may be
Laser Ablation Systems placed in catheters that have a closed-loop
coolant flow to prevent charring and optimize
Thermal ablation involves the physical proper- light penetration into the tissue. Usually, the
ties of the tissue such as the thermal conduc- laser light is in the near infrared portion of the
tivity and the heat capacity. Laser-induced electromagnetic spectrum.
ablation also relies on the optical properties of
the tissue (including absorption and scattering),
coupled with the characteristics of the laser
PhotoMedex
light (including wavelength, energy, power). A The PhotoMedex diffuser (PhotoMedex, for-
medical laser system provides a strong repro- merly SLT, Montgomeryville, PA) is an optical
ducible source of monochromatic light that can fiber that includes a light diffusing segment
be delivered in a controlled fashion. Ablation (active diffusing portion) that ranges from 1 to
techniques either use lenses to focus light to 4cm. The diffusing tip is shown in Figure 7.15.
either a broad or a pinpoint beam, or use The fiber's overall length is either 3.5 or 12m.
7. Devices, Equipment, and Operation of Ablation Systems 87

It is intended for tissue coagulation with wave-


lengths of light from 980 to 1064 nanometers
(nm). The power delivered to the fiber must be
less than 2 W per em (maximum 6 W), that is, no
more than 4 W to a 2-cm tip or 6 W to a 3-cm
or 4-cm tip. Since the fiber's tip is both blunt
and flexible, it is used in conjunction with a
more rigid catheter that is designed for pene-
tration into tissue. This interstitial cooling
catheter, available from Somatex, Inc., is avail-
able in a version that can be cooled with flowing
water (-22°C); this allows up to 12W per cen-
timeter to create larger lesions.
The PhotoMedex CL MD/220 100W medical
FIGURE 7.15. PhotoMedex (formerly SLT) laser laser system is based on a neodymium: yttrium-
fiber. This 1.5-cm diffusing tip provides an extended aluminum-garnet (Nd:YAG) continuous laser
light source for tissue ablation. The fiber disperses emitting at 1064nm, and is shown in Figure 7.16
light radially out along the final length of the tip. The
(8). It is a free-standing system that is internally
fiber is placed into tissue itself within a catheter for
added mechanical support and penetration. The air cooled; it can deliver up to 100W (other CL
catheters can be cooled by a closed-loop flow of MD series versions provide 25,40, and 60W).
water to prevent tissue char that can impede the pen- The primary displays of the main control panel
etration of near infrared light into tissue. (Photo (also shown in Fig. 7.16) show the laser power
printed with permission from PhotoMedex.) and exposure duration. The laser can be run in

A
FIGURE 7.16. PhotoMedex (formerly SLT) laser sys- panel (B) are the power delivered to the tissue and
tem (A). This is a neodymium:yttrium-aluminum- the duration of the exposure. In the figure, 6 W of
garnet (Nd:YAG) laser operating at lO64nm. For power have been delivered over 75.5 seconds. (Photo
interstitial thermal ablation with the laser, the pri- printed with permission from PhotoMedex.)
mary elements of the display on the main control
88 p.R. Morrison and E. vanSonnenberg

a continuous wave (CW) mode or pulsed


model; the CW mode delivers power continu-
ously with the press of a foot pedal; the pulsed
mode provides a shuttering of the light in set
limited exposures from 0.1 to 90 seconds.

Microwave Ablation Systems


Microwave (MW) ablation systems radiate
electromagnetic radiation into tissue where it is
absorbed and transformed into heat to ther-
mally coagulate the target. The applicator is an FIGURE 7.18. Alfresa Pharma Corp. microwave gen-
antenna that emits in the microwave region of erator. The main control panel displays the user-
the electromagnetic spectrum. The antenna selected parameters for the ablation. These include
provides a complete electrical circuit at the tip, the output power, exposure duration, and the disso-
and thus there is no requirement for grounding ciation parameters mentioned in the text. Here, the
pads to be placed on the patient. ablation cycle is set for a duration of 40 seconds at
70W with a dissociation current of 15mA lasting 15
seconds. Also, the generator can power two probes
Azwell simultaneously. (Photo printed with permission from
Alfresa Pharma Corp.)
The Microtaze AZM-520 (Azwell, Inc., Osaka,
Japan) MW ablation system operates at a fre-
quency of 2450 MHz, and provides power ranging between 15 and 25 cm. It is possible to
output up to now (9). The probes (antennae) activate two antennae simultaneously for
are available in a variety of designs for use in added tumor coverage during a single applica-
open, percutaneous, laparoscopic, and endo- tion of energy.
scopic procedures. An example of the tip of a The main control panel of the AZM-520 is
probe for interstitial use is shown in Figure 7.17. shown in Figure 7.18. It is divided into sections:
The probe tips are needle-like to traverse coagulation, dissociation, mode, and memory.
tissues for deep applications; they range from The primary intraprocedural controls are
1.6 to 2 mm in diameter with overall lengths through the power setting and the duration of
exposure, both of which are displayed in the
coagulation section of the panel. Power output
and duration can be adjusted. Separately, the
system provides the dissociation feature, which
establishes a polarity on the antenna that is
intended to electrically attract water molecules
to the probe to induce tissue hydration to
reduce unwanted tissue adherence to the
probe. The dissociation current and cycle time
are set by the user. Separately, a selection
between the normal and auto-repeat modes is
made. The normal mode establishes a preset
FIGURE 7.17. Alfresa Pharma Corp. needle-type
cycle of coagulation and dissociation with a
microwave antenna. The tip of the microwave probe press of the control foot switch; the application
designed for interstitial use is needle-like for tissue of energy ceases when the switch is released.
penetration, and ranges between 1.6 and 2 mm in The auto-repeat mode repeats a number of
diameter. (Photo printed with permission from cycles of preselected outputs without having to
Alfresa Pharma Corp.) maintain pressure on the foot switch. Values for
7. Devices, Equipment, and Operation of Ablation Systems 89

ablation settings can be saved in the memory they are affixed. Through each cannula is
via buttons in the memory section. Sample set- extended a stylette that curves into a 3- (or 4-)
tings are 70W power, 40-second duration, and cm circle on deployment. Each stylette is actu-
15 seconds at 15 rnA dissociation current. ally the antenna for the microwave deposition.
The three circular stylettes together define a
sphere that encompasses the tissue to be
Vivant treated.
The control panel for the VivaWave genera-
The VivaWave microwave generator (Vivant tor is shown in Figure 7.20. The primary feed-
Medical, Mountain View, CA) operates at a back from the system to the user is a visual
source frequency of 915MHz (10). It can display of the elapsed time at a given power.
provide up to 60W of power. The VivaWave is The power output is set by the user for a given
used in conjunction with Vivant's VivaTip or energy deposition-it can be adjusted up to
VivaRing probes. These are shown in Figure 7.19. 60 W in increments of 5 W. There is also a pair
The VivaTip is a 14G needle-type antenna that of control buttons and a display for setting the
can be inserted directly into tissue. It can be duration of exposure from 0-30min in Imin
used as a single probe, or, it can be used in a increments. Cable connections are made on the
configuration together with 2 other probes front of the device. The front panel provides
simultaneously. The multiple probe configura- a visual cue (light), and there is an audible
tion is aligned by a handheld template for tone heard when energy is sent through the
keeping the three probes at a fixed distance probe.
from one another. This is intended to optimize
the volume of ablation to the target volume.
The user can select from three choices for inter- Focused Ultrasound Systems
probe distance. The VivaRing is an array-type
device for microwave delivery to tissue. The Focused ultrasound (FUS) ablation of soft
array is established in tissue after the insertion tissue is distinct from the techniques described
and positioning of three 14 G cannulae; these above in that it is a noninvasive transcutaneous
cannulae are held together at a fixed distance application of energy used to create volumes of
from each other by a common handle to which ablation. Thus, it does not use a probe as an

A B
FIGURE 7.19. Vivant Medical needle-type VivaTip additional probes, as shown, for larger ablation
microwave probe in a multi-probe configuration (A) volumes. The VivaRing provides an array of anten-
and the array-type VivaRing Atom microwave probe nae that is deployed interstitially and intended to
(B). The VivaTip is designed for direct penetration encircle the volume to be ablated. (Photo courtesy
through tissue and can be used individually or with of Vivant Medical, Inc.)
90 P.R. Morrison and E. vanSonnenberg

FIGURE 7.20. Vivant Medical VivaWave microwave the array-type probes require multiple generators.
generator. The main control panel allows the user to The primary feedback to the user is the display of
adjust power (up to 60W) and duration (up to the elapsed time during energy deposition. (Photo
30min) for the ablation. Multiple needle-probes or courtesy of Vivant Medical, Inc.)

applicator, but rather uses a transducer placed also shown in Figure 7.21. Up to this point in
in contact with the skin. Also, FUS is based on this chapter, there has been no discussion of
tissue heating by the transmission and absorp- specific radiologic imaging techniques to plan
tion of high-frequency ultrasonic waves instead or monitor therapy (again, the chapter is an
of electromagnetic energy. The user ablates a overview of the operation of each of the
tumor by covering the targeted volume with systems). However, MRI is integral to the oper-
multiple abutting ellipsoids of coagulation. ation of the ExAblate 2000 system. At all stages
of the ablation process, MRI (a) offers soft
tissue visualization for pre- and postprocedural
InSightec-TxSonics
assessment, and (b) provides quantitative
The ExAblate 2000 (InSightec-TxSonics, thermal mapping of the heating process.
Dallas, TX) FUS ablation system features a The main control panel of the system is
multi-element phased array ultrasound trans- shown in Figure 7.22. The MRI images them-
ducer. A sample transducer is shown in Figure selves help visualize the tumor and establish a
7.21. This multi-element phased array provides treatment plan with the outlining of the volume
the user with the ability to adjust the volume of to ablate. As determined by the planning,
treated tissue per sonication (range 2 x 2 x parameters are selected at the outset of abla-
4mm3 to 10 x 10 x 35 mm3) , as well as the depth tion. For each sonication, these include acoustic
of penetration from a single position outside power (sample: 70W), duration (sample: 20
the body (range 5-20cm). With the patient seconds); duration of cooling in between
lying atop the device, the system can move the sonications (sample: 85 seconds); dimensions
transducer in multiple dimensions to ablate a (sample: 28mm long axis x 6mm diameter
lesion. The transducer is contained in a sealed ellipsoid), and the target temperature (sample:
water-bath that is in contact with the patient for 85°C). Subsequently, spatial coordinates of the
effective transmission of the ultrasound waves sonications consistent with the plan are identi-
into tissue. The operating frequency is centered fied. During the procedure, feedback to the user
on approximately 1.61 MHz. includes the average temperature achieved in
The transducer is incorporated into a modi- each sonication, the number of sonications per-
fied magnetic resonance imaging (MRI) table formed, the number remaining, and the total
7. Devices, Equipment, and Operation of Ablation Systems 91

A B
FIGURE 7.21. InSightec ultrasound transducer. A be adjusted to target tissue at a depth, with individ-
multi-element phased array transducer (A) is inte- ual sonications. Multiple overlapping sonications are
grated into an MRI table (B). The patient is placed used to ablate a targeted volume. (Photos printed
atop the table and the location of the transducer can with permission from InSightec.)

B
FIGURE 7.22. InSightec console. The main control tissue. A cross-hair indicates the location of the
console for the ablation system (A) resides to the left current sonication. In this case, 70 W of focused
of the MRI scanner console (A). Salient features of energy are delivered for 20 seconds; the tissue is
the ablation and monitoring are shown in the sample allowed to cool for 85 seconds before the next
screen (B). Here, an ovoid outline of a target in one sonication. (Photos printed with permission from
MRI scan plane is seen. Individual sonications are InSightec.)
recorded (circles) as the energy is deposited into
92 p.R. Morrison and E. vanSonnenberg

energy delivered to tissue. Further, superim- References


posed on the MRI images is a cumulative visual
1. Instructions for Use 921-91-003 Rev. B. and 460-
record of the ablation associated with each son- 92-001 Rev. F. Radionics, Burlington, MA.
ication for comparison with the original treat- 2. Instructions for Use. Boston Scientific Corpo-
ment plan. ration-Oncology, Natick, MA.
3. User's Guide and Service manual Ref 160-
102025, RITA Medical Systems, Mountain
Conclusion View, CA.
4. http://hittmethod.com, Berchtold, GmbH, Tut-
This chapter higWights the operation of a tlingen, Germany.
number of medical devices for thermal abla- 5. User's Manual. Ref: 991.027 and 991.0287/2002.
tion. It reveals the scope of the thermal modal- Celon AG Medical Instruments, Berlin,
Germany.
ities available to physicians to induce tissue
6. Operator's Manual. Galil Medical, Westbury,
necrosis: from heating at +100°C to a NY.
-180°C freeze; from passing an electrical 7. http://www.endocare.com. Endocare, Inc.,
current to depositing invisible photons; from Irvine, CA.
manipulating the placement of an antenna into 8. Operations and Maintenance Manual for the
tissue to the remote sonication of a tumor from CL MD Series Laser. PN 0800-0370. Rev.
a computer console. Ten ablation systems have 5.11/94. PhotoMedex formerly SLT, Mont-
been reviewed. Readers should consider these gomeryville, PA.
systems as representative of the ablation mech- 9. Operation Manual Microtaze AZM-520,
anisms, and interested individuals are encour- Azwell, Inc. Osaka, Japan.
aged to investigate each for themselves and to 10. VivaWave Microcoagulation System User
Manual, PN0339 Rev C. Vivant Medical, Moun-
understand the principles behind the devices to
tain View, CA.
better assess their use and implementation. 11. Personal communications. InSightec-TxSonics,
Dallas, TX.
Section III
Imaging for Tumor Ablation
8
Intraoperative Ultrasound-Guided
Procedures
Robert A. Kane

While the continuous development and im- probes tend to be smaller than conventional
provements in tumor ablation techniques and abdominal ultrasound probes. Since the probe
equipment have made the percutaneous non- can be positioned directly on the target organ,
surgical approach ever more successful, even higher scanning frequencies can be utilized suc-
for larger lesions, there is still a need for abla- cessfully as there is much less attenuation of the
tive treatments in the operating room. In some sound beam than when scanning through the
practices, based on local referral patterns, the skin and subcutaneous fat and other interven-
surgical approach to tumor ablation may be ing tissues. The probes designed for intraoper-
predominant, either via laparoscopy or open ative abdominal use are linear, curvilinear, or
laparotomy. In other settings, additional tumor phased array electronic probes that have the
sites may be encountered during a planned capacity both for gray scale imaging as well as
surgical excision that can be efficiently treated spectral and color Doppler scanning. Some
intraoperatively with a variety of ablative manufacturers offer probes with a choice of
approaches. Finally, some techniques, such as multiple frequencies (5-7.5 MHz), while others
cryoablation, may be best performed intra- utilize broadband technology to display a
operatively, particularly when treating large range of frequencies from near to far field,
tumors with 5- to lO-mm diameter cryoprobes. and for abdominal uses ranging from 3 up to
Therefore, a review of intraoperative ultra- 10-12 MHz.
sound (lOUS) scanning and guidance tech- Because of the sophisticated electronics and
niques for ablation is appropriate. This chapter the encasing plastic housing and rubber mem-
discusses the optimal approach to intra- branes over the transducer, heat sterilization
operative and laparoscopic ultrasound (LUS) via autodaving is not possible. Some of the
scanning techniques, equipment, technical earliest intraoperative probes were designed
preparations, and methods for guidance of for direct sterilization using techniques such as
interventional techniques. Since most of the ethylene oxide gas sterilization or prolonged
intraoperative tumor ablations involve lesions immersion in glutaraldehyde. However, other
in the liver, we focus the discussion on hepatic manufacturers prohibited gas sterilization
ultrasonography and guidance. because of concerns of possible damage to the
transducer. In addition, with heightened envi-
ronmental concerns more recently, many hos-
Equipment pitals and operating suites no longer allow the
use of such agents as ethylene oxide or glu-
All of the major ultrasound manufacturers taraldehyde. Therefore, the method of choice
have specifically designed probes for open for maintaining sterilization in the operative
intraoperative ultrasound uses (Fig. 8.1). These field is the use of specifically designed sterile

95
96 R.A. Kane

with a long sterile sleeve that can extend over


the electronic cord to connect the transducer to
the ultrasound scanner console (1).
Several, but not all ultrasound companies
also manufacture laparoscopic ultrasound
probes that are designed to fit through a stan-
dard 10- to ll-mm laparoscopy port. These are
either linear or curvilinear transducers. They
are approximately 2 to 4cm in length and are
mounted on a 30- to 40-cm-Iong shaft. The very
FIGURE 8.1. Intraoperative ultrasound (IODS) first of these systems was entirely rigid,
probes. Various linear and curvilinear, end-fire, but this proved impractical, since good contact
and side-fire probes are available for intraoperative could not be maintained easily on the curved
use. The side-viewing probes are most optimal surfaces of the abdominal organs. Currently
for imaging the liver. available systems offer flexibility of the scan-
ning tip, with systems similar to those used
on endoscopes (Fig. 8.2). These systems allow
probe covers that are individually manufac- either two-way flexion and extension maneu-
tured to fit snugly over the specific size and verability or four-way maneuverability, includ-
shape of the intraoperative ultrasound probe. ing left and right deflection, as well as flexion
Acoustic coupling is required between the and extension (2). As with open abdominal
probe and the probe cover, which is most ultrasound probes, pulsed Doppler and color
optimally accomplished by using sterile gel or flow imaging are available, and the systems also
possibly sterile saline. A snug fit must be main- offer either multiple frequencies or broadband
tained over the transducer surface so that no air technology. Covers for laparoscopic probes
bubbles are present between the transducer require a long, thin, sterile sheath that must be
and the target organ, since these will cause arti- applied tightly to the shaft of the laparoscopic
facts due to acoustic shadowing. Most of the probe to fit easily through the laparoscopic port
probe covers also are manufactured complete without tearing. Once again, acoustic coupling

B c
FIGURE 8.2. Laparoscopic ultrasound (LDS) probes. (A) Linear array probe in flexion. (B) Curved array
probe demonstrating left deflection. (C) Close-up of curved array probe demonstrating the biopsy port
(arrow).
8. Intraoperative Ultrasound-Guided Procedures 97

is required at the transducer interface, usually can be obtained either by increasing compres-
accomplished by placing sterile saline in the sion of the anterior liver and placing the focal
sheath prior to insertion of the probe (1). zones more deeply in the liver, or by scanning
from the lateral or deep liver surfaces when
necessary.
Scanning Technique Using laparoscopic ultrasound, the technical
challenges to obtaining a complete scan of
While it is essential to provide acoustic cou- the liver are greater. Often, the laparoscopic
pling between the surface of the transducer ultrasound probe must be exchanged with the
and the overlying sterile sheath using either laparoscope, and scans must be obtained from
sterile gel or sterile saline, the natural moisture more than one port site (2).1Ypically, a midline
within the body cavity is usually sufficient for or periumbilical port may provide good access
good acoustic coupling between the ultrasound to the left lateral segment, but the falciform lig-
probe and the liver or other intraabdominal ament may impede positioning of the probe on
organs. If the surfaces are somewhat dry, sterile the right side of the liver. In other patients,
saline can be applied over the organ surface a periumbilical or midline port may provide
to improve acoustic coupling. For maximal better access to the right lobe, with associated
detection of occult liver lesions, a systematic difficulties scanning to the left of the falciform
approach to scanning the liver should be fol- ligament. A right subcostal port may be helpful
lowed carefully (3). We prefer to scan from the to image the right lobe; care must be taken
anterior surface of the liver that is relatively that this port is not placed too close to the tip
smooth, rather than the deep surface, which has of the right lobe to leave room to maneuver the
numerous fissures and undulations, and may probe over the anterior surface of the liver. As
present difficulties in maintaining good contact with open intraoperative scanning, a consistent,
with the liver. Side-viewing probes are pre- planned approach to scanning the liver is
ferable to allow scanning of the entire liver important to obtain a complete study. Our
surface. Our scanning convention is to begin approach is to begin either to the right or left
with the cephalad-lateral aspect of the left lobe of the falciform ligament and fan the probe
(segment II). The probe is positioned in a trans- across the dome of the liver from the medial to
verse orientation and scans are performed lateral position, and then withdraw the probe 2
slowly in a cephalocaudad direction until the to 4cm (the length of the probe surface) and
caudal edge of the liver is reached. This maneu- repeat the scans again from medial to lateral,
ver is then repeated by moving the probe withdrawing and scanning until the entire liver
toward the right by the width of the transducer has been imaged from cephalad to caudal
and repeating a cephalocaudad sweep. In this margins. This is much more tedious and time-
fashion, moving left to right, the entire liver can consuming than with open intraoperative scan-
be scanned with approximately five to eight ning, since the probe lengths are substantially
cephalocaudad sweeps, taking approximately 5 smaller for laparoscopic ultrasound systems.
minutes for a complete scan (4). Each succes- Consequently, a complete scan of the liver using
sive series of sweeps should slightly overlap the the laparoscopic approach takes at least 10 to
preceding one to ensure coverage of the entire 15 minutes to accomplish.
liver. Numerous studies have demonstrated the
The optimal frequency for liver imaging is superiority of intraoperative and laparoscopic
5MHz, since the attenuation of soft tissue in the ultrasound imaging over preoperative imaging
liver may be too great at higher frequencies, studies for maximal liver lesion detection (Fig.
thereby compromising imaging of the deep 8.3). The spatial resolution is capable of depict-
portions of the liver. With fatty infiltration, ing cystic structures as small as 1 to 3 mm and
even at 5 MHz, complete penetration may be a solid nodules as small as 3 to 5 mm. Studies have
problem, but if a lower frequency option is not demonstrated improved liver lesion detection
available, satisfactory images of the deep liver of 20% to 30%, compared to standard com-
98 R.A. Kane

puted tomography (CT) and magnetic reso-


nance (MR) techniques (5,6), and increased
detection of 10% to 15 % compared to CT arte-
rial portography (7). More recently, develop-
ment of multidetector CT scanning, as well
as improvements in MR technology resulting
in decreasing scan speeds have markedly
improved detection of small liver lesions,
particularly with multiphase contrast-enhanced
scans. There are no recent studies to compare
the efficacy of intraoperative imaging to these
latest high-speed scanning techniques, and,
undoubtedly, fewer additional lesions will prob-
ably be detected by intraoperative scanning, A

B
FIGURE 8.4. LUS images in a patient with hepato-
cellular carcinoma (HCC). (A) A 2.5-cm HCC in a
cirrhotic liver. (B) Unsuspected 5.6-mm HCC
A nodule. Two unsuspected nodules were identified at
LUS, not seen on preoperative imaging.

but some additional lesions will still be detected


and may be highly influential on the selec-
tion of appropriate intraoperative therapies
(Fig. 8.4). Therefore, a compulsive systematic
assessment of the entire liver is still of con-
B siderable importance. In fact, detection of
FiGURE 8.3. IOUS liver in a patient with breast additional unsuspected lesions often is an event
metastases. (A) A 3-mm hepatic cyst (arrow). (B) A that results in a tumor ablation technique being
6-mm echogenic metastasis (arrow), not seen on pre- undertaken in the operating room either
operative imaging. Three additional metastases were instead of, or in addition to, the planned surgi-
identified at IOUS. cal tumor resection (8).
8. Intraoperative Ultrasound-Guided Procedures 99

Ultrasound Guidance
Techniques
If additional unsuspected lesions are encoun-
tered during scanning, ultrasound-guided
biopsy and immediate frozen section analysis
of the biopsy can be performed to confirm the
presence of additional tumor sites. At open
intraoperative ultrasonography, this can be
readily performed using the free-hand tech-
nique (9). The lesion is positioned under the
ultrasound probe and a needle is advanced
along the midline of the long axis of the trans- FIGURE 8.6. Electronic biopsy guidance. The hypo-
ducer, such that the entire length of the needle echoic tumor is positioned within the electronic
can be imaged. This allows precise changes cursors, which can be used to guide placement of
in angulation of the needle that can be placed biopsy needles or ablation devices.
accurately within 1 to 2mm of the target. We
utilize automated is-gauge core biopsy needles
to obtain intraoperative biopsies. Occasionally,
length of the laparoscopic ultrasound probe
a lesion may be so deeply seated that the free-
(Fig. 8.2C), but this is a somewhat cumbersome
hand technique is difficult. Fixed electronic
device to use, and it is expensive.
biopsy guides are not available for most intra-
An alternative approach to laparoscopic
operative ultrasound probes, but there is an
ultrasound-guided biopsy is to puncture
alternative solution that we have used success-
through the anterior abdominal wall, using
fully for deep, difficult needle placements.
a long laparoscopic biopsy needle, and then
This technique utilizes an end-fire endorectal
position the needle immediately adjace~t to the
prostate probe covered with a sterile sheath
laparoscopic ultrasound transducer, usmg the
(Fig. 8.5). Over this sheath, a sterile biopsy
real-time images to adjust the angulation and
guide can be positioned and a precise electronic
depth of the biopsy needle (2). This approach
depiction of the path of the biopsy needle can
is feasible with lesions that are of moderate
then be utilized for accurate needle positioning
size and not too deeply placed within the liver.
(Fig. 8.6), even at 15 cm depth.
However, attempting to biopsy small, unsus-
Biopsy guidance during laparoscopic ultra-
pected lesions detected during laparoscopic
sound imaging is considerably more difficult
ultrasound can be quite difficult, since these
and problematic. Most laparoscopic ultrasound
additional lesions tend to be quite small, usually
systems lack electronic biopsy guidance
less than 1 to 2 cm in size.
systems, although there is one system that
The same techniques used to biopsy liver
allows passage of a biopsy guide through the
lesions are also used to accurately place needles
and probes for tumor ablation (10). With
alcohol ablation or hyperthermia techniques
such as radiofrequency, microwave, and laser
ablation, the size of the needles and probes
approximates 16 to 21 gauge, and hen.ce free-
hand ultrasound guidance is quite feaSIble and
accurate (Fig. 8.7). Once again, utilizing an en~­
FIGURE 8.5. Curvilinear endoluminal probe with fire prostate-type probe with an electroUlc
biopsy guide. The biopsy needle or ablation probe biopsy guidance system is useful for accurate
can be positioned in the trough of the biopsy guide placement of probes and needles into small,
for precise intraoperative placement. deeply situated lesions.
100 R.A. Kane

A B

FIGURE 8.7. Ethanol ablation. (A) A 1.3-cm nodule


(calipers) of hepatocellular carcinoma. (B) A 20-gauge
needle (small arrows) being advanced with free-hand
technique toward the HCC nodule (large arrow). (C)
Following injection of absolute alcohol, the HCC
c nodule is obliterated by hyperechogenic microbubbles.

For cryoablation, the size of the cryoprobes For all intraoperative or laparoscopic abla-
tends to be somewhat larger, ranging from 3 to tion techniques, ultrasound is imperative to
5mm and even 10mm in diameter. These larger ensure proper placement of the probes or
probes are more difficult to place under direct needles, as well as to monitor the actual ablation
real-time guidance. If the lesion is sufficiently procedure. For optimal therapeutic efficacy, it
large, the probes can be placed manually with is essential that the ablation margin extends
intermittent ultrasound imaging to confirm beyond the borders of the tumor into the sur-
accurate placement, or alter the course and rounding normal liver parenchyma. Ideally, one
depth of penetration as needed (11). Another seeks to create a I-em margin of normal liver
approach is to use smaller, 18-gauge needles around all borders of the tumor (13) to minimize
for precise placement, and then place the local recurrence due to inadequately lethal tem-
larger cryoprobe alongside the guiding needle peratures at the periphery of the mass.
in tandem, or by the Seldinger technique, using With cryoablation, the iceball is sharply
an 18-gauge needle, guidewires, and dilators defined and precisely imaged as an echogenic
(12). Whatever method is used, precise place- curvilinear freeze front, behind which there
ment is essential to ensure adequate freezing of is complete acoust~c ~hadowing. Studies have
the entire lesion and the contiguous liver. established that Within a few millimeters of this
8. Intraoperative Ultrasound-Guided Procedures 101

visible freeze front, lethally cold temperatures well as injection of absolute alcohol, acetic
will have produced complete necrosis (14). The acid, or other fluid, such as heated saline. Once
difficulty encountered in cryoablation is that again, the goal is to extend the ablation margin
only the advancing half of the freeze front can into the surrounding liver on all sides of the
be imaged while the deep portion is obscured tumor. However, these techniques usually
by acoustic shadowing. Consequently, for com- produce microbubbles within the ablated tissue
plete monitoring of the cryoablation, images soon after treatment has begun (15). These
must be obtained from the deep surface as well microbubbles cause acoustic shadowing along
as the superficial surface whenever possible. In the deep surface and result in considerable
this fashion, the freeze front can be observed to ultrasound scatter at the superficial and lateral
extend through the tumor into the surrounding margins of the tumor, resulting in difficulty in
liver, and the freezing time can be extended as precise definition of the ablation front. As with
long as required for complete tumor treatment cryoablation, imaging from the opposing deep
(Fig. 8.8). The limitation occurs when the freeze surface may help to overcome the acoustic
front extends to one of the liver surfaces, in shadowing, as long as the treated area does not
which case images can be obtained only from extend to the deep surface.
the opposite surface, and hence a complete One of the limitations of both hyperthermic
sonographic assessment may not be attainable. and cryoablation techniques occurs with
There are also limitations in monitoring the tumors that are contiguous to large arterial or
various hyperthermic ablation techniques as venous structures. The flowing blood in these

A B

FIGURE 8.8. Cryoablation. (A) IOUS image showing two


cryoprobes (arrows) placed within a metastasis from
colorectal carcinoma. (B) Partially developed iceballs
are coalescing, but scans demonstrate residual tumor
anteriorly (arrows) requiring further extension of the
cryoablation for complete treatment. (C) After 12
minutes of freezing, the cryolesion (calipers) completely
encompasses the tumor and extends beyond into normal
liver. c
102 R.A. Kane

TABLE 8.1. Advantages and disadvantages of the intraoperative and percutaneous approaches to tumor
ablation.
Approach Advantages Disadvantages
Intraoperative IODS for maximal lesion detection General anesthesia
Safety-avoidance of bowel, gallbladder, heart, Surgical recovery time at least 1-3 days in hospital
lung Monitoring of ablation margins may be less precise
Safety-eontrol of bleeding Limited access to some tumor sites with
Pringle maneuver to decrease heat sink effect laparoscopic approach
Easy access to tumor sites with open surgery Expensive-I-2 hours of operating room time
Percutaneous Local anesthesia/conscious sedation Less complete lesion detection
Short recovery time-same day discharge Avoidance of critical structures (bowel,
More accurate monitoring of ablation margins gallbladder, heart, lung) can present problems
with contrast-enhanced CT, MRI, or DS Difficult to access some lesions
Repeat treatments feasible Cannot restrict blood flow to decrease heat sink
Less expensive effect
Possible tumor seeding of percutaneous access
tract

IODS, intraoperative ultrasound.

large vessels can dilute and carry off sufficient rent disease. Actuarial survival results from
heat or cold to cause the temperatures in the various ablation procedures appear to be
contiguous portion of tumor to remain in the showing comparable results (17-19). Therefore,
sublethal zone. This is probably a frequent it is reasonable to suggest that percutaneous
cause of local failure of various ablation techniques for tumor ablation in the liver
techniques. In the intraoperative approach are preferable to intraoperative approaches in
to liver tumor ablation, a Pringle maneuver can general.
be performed by placing an atraumatic occlu- The most mature survival data are for surgi-
sive clamp across the vascular pedicle to the cal resection of liver tumors. The experience
liver, thereby restricting hepatic arterial and with the various ablation techniques is not as
portal venous inflow. The effects of this mature, nor have there been sufficient prospec-
maneuver can be assessed by intraoperative tive randomized trials comparing resection to
ultrasonography, particularly using color flow various forms of tumor ablation (16,20). For
imaging to demonstrate the reduction of blood this reason, there will still be a demand for
flow. intraoperative approaches to tumor ablation,
particularly when unexpected findings are
encountered at the time of a planned surgical
resection.
Conclusion
There are advantages and disadvantages
to both the intraoperative and percutaneous References
approaches to tumor ablation (16-18) (Table
8.1). However, at least in the liver, for which we 1. Sammons LG, Kane RA. Technical aspects
of intraoperative ultrasound. In: Kane RA, ed.
have the most extensive experience and sur-
Intraoperative, Laparoscopic, and Endoluminal
vival data, all of the approaches to tumor treat- Ultrasound. Philadelphia: Churchill Livingstone/
ment, including surgical resection, are relatively Saunders, 1999:1-11.
ineffective over time. The most successful sur- 2. Kane RA. Laparoscopic ultrasound. In: Kane
gical series have shown approximately 30% RA, ed. Intraoperative, Laparoscopic, and Endo-
five-year survival, and a significant portion of luminal Ultrasound. Philadelphia: Churchill
those five-year survivors are alive with recur- Livingstone/Saunders, 1999:90-105.
8. Intraoperative Ultrasound-Guided Procedures 103

3. Kane RA. Intra-operative ultrasound. In: Meire 11. McPhee MD, Kane RA. Cryosurgery for hepatic
HB, Cosgrove DO, Dewbury KC, Farrant P, tumor ablation. Semin Intervent RadioI1997;14:
eds. Clinical Ultrasound: A Comprehensive 285-293.
Text: Abdominal and General Ultrasound. 2nd 12. Littrup PJ, Lee FT, Rajan D, Meetze K, Weaver
ed. London; Churchill Livingstone, 2001:143- D. Hepatic cryotherapy: state-of-the-art tech-
164. niques and future developments. Ultrasound Q
4. Kruskal JB, Kane RA. Intraoperative ultrasound 1998;14(3):171-188.
of the liver. In: Kane RA, ed. Intraoperative, 13. Ravikumar TS, Kane RA, Cady B, et a1. A 5-year
Laparoscopic, and Endoluminal Ultrasound. study of cryosurgery in the treatment of liver
Philadelphia: Churchill Livingstone/Saunders, tumors. Arch Surg 1991;126:1520-1523.
1999:40-67. 14. Onik G, Kane RA, Steele G, et a1. Monitoring
5. Machi J, Isomoto H, Kurohiji T, et a1. Accuracy hepatic cryosurgery with sonography. AJR 1986;
of intraoperative ultrasound in diagnosing liver 147:665-669.
metastases from colorectal cancer: evaluation 15. DeSanctis JT, Goldberg SN, Mueller PRo Per-
with postoperative follow-up results. World J cutaneous treatment of hepatic neoplasms: a
Surg 1991;15:551-556. review of current techniques. Semin Intervent
6. Clarke Mp, Kane RA, Steele GD, et a1. Prospec- RadioI1997;14(3):255-284.
tive comparison of preoperative imaging and 16. Simonetti RG, Liberati A, Angiolini C, Pagliaro
intraoperative ultrasonography in the detection L. Treatment of hepatocellular carcinoma: a sys-
of liver tumors. Surgery 1989;106:849-855. tematic review of randomized controlled trials.
7. Soyer P, Levesque M, Elias D, Zeitoun G, Roche Ann OncoI1997;8:1l7-136.
A. Detection of liver metastases from colorectal 17. Livraghi T, Bolondi L, Buscarini L, et aI., and
cancer: comparison of intraoperative US and CT the Italian Cooperative HCC Study Group. No
during arterial portography. Radiology 1992;183: treatment, resection and ethanol injection in
541-544. hepatocellular carcinoma: a retrospective analy-
8. Kane RA, Hughes LA, Cua EJ, et a1. The impact sis of survival in 391 patients with cirrhosis.
of intraoperative ultrasonography on surgery J Hepatol 1995;22:522-526.
for liver neoplasms. J Ultrasound Med 1994;13: 18. Korpan NN. Hepatic cryosurgery for liver metas-
1-6. tases: long-term follow-up. Ann Surg 1997;225:
9. Lee RA. Kane RA, Lantz EJ, Charboneau Jw. 193-201.
Intraoperative and laparoscopic sonography of 19. Goldberg SN, Lazzaroni S, Meloni F, et a1. Hepa-
the abdomen. In: Rumack CM, Wilson SR, Char- tocellular carcinoma: Radio-frequency ablation
boneau JW, eds. Diagnostic Ultrasound, vol 1. St. of medium and large lesions. Radiology 2000;
Louis: Mosby, 1998:671-699. 214:761-768.
to. Patterson EJ, Scudamore CH, Buczkowski AK, 20. Rush BJ, Koneru B. Locoregional techniques in
Owen DA, Nagy AG. Radiofrequency ablation the treatment of hepatic tumors. J Surg Oncol
in surgery. Surg Technol Int 1997;6:69-75. 1997;64:259-261.
9
Computed Tomography Imaging for
Tumor Ablation
Thierry de Baere

Computed tomography (CT) affords the best current ablation systems are unable to confirm
visualization of all organs in the body, simulta- complete tumor eradication at the time of treat-
neously depicting air, soft tissue, and bones on ment. The treated organ, therefore, requires
the same scan. This ability makes CT an ideal follow-up imaging to ascertain whether abla-
imaging modality for tissue ablation, as it tion of the targeted tumor was efficient, as
enables the physician to target any type of well as to detect new tumors and treatment
organ accurately and to avoid inadvertently complications.
puncturing others along the needle path.
However, in the past, as the duration of image
acquisition and reconstruction was long with
CT, this time-consuming aspect appeared incon-
Image Guidance
venient for tissue ablation. Recent improve-
ments in computerized data management have
Indications
transformed CT into almost a real-time imaging The first step in the ablation procedure is to
technique, and even more recently, the advent guide the ablation instrument (the probe, the
of multislice CT has made imaging of a volume needle, the fiber, the electrode) toward the tar-
in a single acquisition feasible, thereby improv- geted tumor. Imaging used for guidance should
ing the scope of CT guidance for tissue ablation. clearly depict the ablation instrument and the
CT can now be used throughout tissue ablation targeted tumor, so that the instrument is posi-
for guidance, monitoring, and follow-up. tioned accurately within the tumor. Computed
Image guidance is critical when ablating a tomography is mandatory for guidance when it
tumor, because accurate positioning of the is the only technique capable of imaging the
ablation instrument in the targeted tumor is a targeted tumor. Such is the case for most lung
key factor for treatment efficacy. Moreover, and bone tumors, given that fluoroscopic guid-
accurate guidance avoids nontargeted struc- ance is not accurate enough, and that interven-
tures or organs located close to or in the path tional magnetic resonance (MR) is far from
of the tumor. commonplace today. Such is also the case for
Image monitoring is also important. Ablation some soft organ tumors that are not well
should be tailored to the tumor target volume defined by ultrasound (US). Computed tomog-
to avoid incomplete treatment, recurrence, and raphy can be useful when the tumor is depicted
the need for re-treatment. Injury to neighbor- by US, but is virtually inaccessible for puncture
ing tissue and collateral damage also can be with US on account of the location, or because
evaluated in real time. gas or bones obstruct the imaging window or
Follow-up imaging is mandatory after tumor needle tract. For example, tumors that are
ablation because monitoring techniques and seated high in the liver are sometimes inacces-

104
9. Computed Tomography Imaging for Tumor Ablation 105

sible to puncture under US, whereas CT guid- and time must be limited. The quality of the
ance allows access via the transthoracic route, image needed for guidance is not the same
which is impossible with US guidance due to air as that needed for diagnostic purposes, and
in the lung that obstructs the imaging window. usually is not as good.
Computed tomography is also preferred when Scanning time should be as short as possible
a safe pathway to the targeted tumor cannot be for image reconstruction. Consequently, almost
imaged with US, for example, when a hollow real-time CT imaging, or fluoro-CT, is now pre-
organ containing air is very close. Finally, com- ferred. Continuous fluoro-CT with systems able
bining CT and US guidance sometimes can be to produce at least eight images per second with
useful. Computed tomography can be used to a reconstruction time of less than 0.2 second
guide the ablation instrument toward the tar- allows almost real-time guidance. This has
geted organ along a safe tract that cannot be been demonstrated to shorten the duration of
easily depicted with US, and once the organ has the puncture procedure, and to improve the
been reached, US can then be used to guide the accuracy of tumor targeting, demonstrated
instrument in real time into an ill-defined tumor by an increase in the sensitivity and the nega-
on CT. tive predictive value when used for biopsy
When tumor visualization and accessibility (2).
for puncture are equivalent under both US and If imaging time is maintained at a minimum,
CT guidance, the technique of choice is that and certain precautions such as milliampere
preferred by the operator or according to the reduction for scans to evaluate needle position,
availability of the equipment. In clinical prac- fluoro-CT does not increase the dose to the
tice today, most liver tumors are treated under patient or the physician (3). However, one of
US guidance because this technique is widely the drawbacks of real-time fluoro-CT is that the
available, the cost is low, angulation possibilities needle is handled by the physician while x-rays
are virtually limitless, and real-time guidance is are being delivered, and this increases the
achievable. Today, with the advent of almost radiation received by the physician's hands that
real-time CT, often called CT fluoroscopy are in, or close to, the imaging field. Special
(fluoro-CT), the time required for needle inser- needle holders have been designed so that the
tion during ablation is short, thus overcoming physician can guide the needle while keeping
the previous drawback with CT. A comparative his/her hands away from the x-ray beam (4,5).
study on a phantom demonstrated no signifi- To further minimize radiation delivered to the
cant differences between US and fluoro-CT in patient and the physician, the "quick-check"
the time needed to guide a biopsy; helical CT technique can be used. This technique uses
guidance was threefold longer (1). brief single-section fiuoro-CT imaging that is
repeated whenever the needle is advanced,
but unlike real-time fiuoro-CT, the needle is
Technique advanced without real-time imaging. The quick-
Some technical requirements need to be em- check technique is accurate and rapid for
phasized. The gantry must be large enough to guiding the needle to the target (6). In practice,
allow the passage of the instrument that has the quick-check technique is adequate for the
been partially inserted in the patient. As some majority of cases. Continuous fiuoro-CT prob-
handles on the instruments are quite long, a ably should be reserved for difficult cases, for
handle-less guiding trocar needle or a flexible example, in moving structures, or when the
device may facilitate placement in the tumor. access for the target is narrow, or finally for the
For practical and sterility reasons, a TV screen actual puncture when the instrument is pene-
is mandatory inside the CT room. It is also trating the targeted tumor.
useful to have a footswitch to use to direct and Multislice CT is even more user friendly for
release the table, and to be able to perform CT puncturing under CT guidance, as it offers one
acquisition inside the room. To minimize radia- or two center rows to image the needle course,
tion to the patient and to the physician, the dose and two lateral ones to ensure that the instru-
106 T. de Baere

ment does not overstep the track in the upper below the tool to ensure that the tip is not over-
or lower imaging planes. The most recent mul- stepping the reference slice.
tislice CT unit with up to 16 rows provides rapid With conventional CT guidance, only tumors
three-dimensional (3D) volumetric imaging to visualized without injection of contrast medium
such an extent that it may be possible in the were punctured, because the time necessary to
near future to guide the needle in all planes perform the puncture was longer than the dura-
with the help of real-time reconstruction. tion of enhancement. Nowadays, with fluoro-
However, this type of software is not capable of CT, a tumor can be punctured during transient
providing oblique multiplanar imaging in real- enhancement, as described in the literature for
time as yet. This is why, even today, a perpen- tumors enhancing from 50 to 130 seconds (8,9).
dicular approach is always preferred when The target lesion should be in the scan plane to
possible, because it is the simplest. If an angled do so. This scan plane and delayed imaging
approach is used, it allows the entire puncture should be determined according to temporal
tract and needle to be imaged in one slice (7). measurement of delay times, based on the pre-
In practice, the procedure for inserting the vious diagnostic CT examination. The physician
ablation instrument begins with a detailed CT should be ready, standing at the side of the
examination of the region. The patient is placed patient clothed in a sterile garment in front of
in the most favorable position for access to the the gantry, poised for the puncture.
entry point. Treatment of a limb tumor may When the puncture cannot be performed
require a different degree of rotation. Securing within gantry angulation possibilities, it will be
the limb with tape or straps can be useful less accurate and more time-consuming. In the
during treatment of bone tumors, because the past, the only alternative was to use the trian-
force required for drilling can cause extremity gulation technique in such situations. Today,
movement that can be bothersome on subse- guidance with a laser goniometer (10) or elec-
quent CT scans. Next, the level of the targeted tromagnetic virtual targeting systems (11) can
slice is highlighted with a laser marker light, be extremely helpful as they heighten accuracy
and the entry point is marked on the skin. A and shorten the duration of the procedure and
metallic marker can be used to delineate the the number of needle passes. However, the
entry point precisely. The anticipated angle of remaining disadvantage of these out-of-plane
the path and depth of the target are determined techniques is the nonvisualization of the com-
according to this entry point using the elec- plete needle on a single image. Electromagnetic
tronic calipers on the CT console. targeting systems are composed of a computer
Once these parameters have been defined, it that is first loaded with a set of reference CT
is often useful to stick a short, small-caliber scans obtained immediately before puncturing
needle in this entry point that can be used for the tumor, and the patient is maintained immo-
local anesthesia if needed. This needle confirms bile thereafter. This set of CT scans encom-
that the entry point and tract angulation are passes the target and the entry point when an
accurate on the subsequent CT scan, before out-of-plane puncture is needed, or provides a
insertion of the ablation instrument using the single reference slice when an in-plane punc-
tandem technique. Then, while the clinician is ture is to be performed. The target and the
advancing the instrument, CT imaging can be entry point are then marked with calipers on
obtained at regular intervals to verify the depth the screen of the targeting system. Before actu-
and direction of the incoming instrument, and ally inserting the needle, a virtual needle path
this depends on the rapidity of image acquisi- can be visualized on the set of CT scans, and
tion and reconstruction. Real-time fluoro-CT multiplanar reconstruction can be performed in
or quick-check imaging can be used, as dis- any direction and angulation. Once this viewing
cussed above. A distal shadow due to a partial- has demonstrated that no vital structures would
volume effect allows the physician to dis- be violated, the puncture can be performed fol-
tinguish the tip of the instrument, but one lowing the direction indicated on the screen of
should always image a slice above and another the guiding system. During the puncture, an
9. Computed Tomography Imaging for Tumor Ablation 107

electromagnetic sensor placed on the proximal the entry point on the patient's skin. The direc-
needle, close to the needle hub or instrument tion of the needle is kept within the laser beam,
handle, allows (with the help of algorithms demonstrating the desired angle while advanc-
similar to those used in global positioning ing the needle for the puncture. The greatest
systems) accurate localization of the needle tip contribution of such a system occurs when per-
in real time, as well as the route chosen by the forming double angulation punctures within
physician, both of which are superimposed over the limits of gantry angulation (10,12).
the reference set of previously loaded images As thermal ablation instruments are rela-
(Fig. 9.1). The electromagnetic targeting system tively large in caliber, the best option is to
has a built-in respiratory phase monitoring access the target organ without traversing any
system that allows the needle to be inserted other structure. When the window for safe
during the same phase of the respiratory cycle access to the target organ is quite narrow, or
as that during which reference images were even virtual, the physician can inject a large
acquired. As the position of the needle tip and volume of saline to widen the path artificially
its future route and position are a "straight pro- by enlarging the anatomic space between two
jection" of the direction of the proximal part of organs. This technique has been reported for
the needle, great care should be taken not to biopsy in the mediastinum (13,14), the retro-
bend the needle, as this will lead to incorrect peritoneum (15), and the pelvis (16). For this
projection of the position of needle tip and technique, first, a small (often 22 gauge), caliber
future path. Using a small-gauge needle with a needle is inserted as deeply as possible in the
bevel can be problematic, since the needle tract narrow tract chosen for insertion. Then, saline
may become curved. Rotating the needle while is manually injected slowly to enlarge the tract
advancing it has been described to minimize and move the untargeted organ away. The
possible curving and deviation. needle is then advanced further and the injec-
The laser goniometer is a laser light unit that tion is repeated until a large-enough area is
can be either attached to the gantry or be sep- created for insertion of the ablation instrument.
arate from it. Angulate coordinates, determined Usually 10 to 30mL of saline are used, but up
on the CT unit from previously acquired slices, to 60mL may be required.
are calculated. Then the laser beam is angulated In high-seated liver tumors that are inacces-
according to these coordinates, and focused on sible under US, access using CT often is diffi-

FIGURE 9.1. View of the screen


of an electromagnetic targeting sys-
tem. The circle on the right frame
represents the location of the
needle tip. The parallel lines on the
left frame represent the future
needle path; the distance to the
target is also shown. Note the two
respiratory sensors on the anterior
abdominal wall used to monitor the
respiratory cycle. (Courtesy of Dr.
Afshin Gangi.)
108 T. de Baere

injected into the pleural cavity. Usually, 100 to


400mL of air are enough to clear the lung from
the needle path. Finally, the lung is separated
from the diaphragm; then the liver can be
accessed through the air-filled pleural space
while avoiding injury to the lung, and repro-
ducing, in a manner akin to the surgical
transpleurodiaphragmatic access described by
surgeons, an efficient way of treating tumors
of the dome of the liver (18). At the end of
the ablation procedure, the needle is exchanged
over a 0.035-inch wire for a 5-French side-
hole catheter to remove air from the pneu-
mothorax, and to expel it through a three-way
FIGURE 9.2. Computed tomography (CT) scan stopcock.
depicts insertion of a cool-tip cluster radiofrequency Treating tumors in proximity to organs that
(RF) needle in a very high-seated segment VII liver are sensitive to heat or cold can cause collateral
metastasis via an air-filled right pleural cavity. The damage to these organs. Such complications
large right pneumothorax has been induced to
have been reported to occur in the colon,
avoid piercing the lung. This pneumothorax will
be expelled after transpleurodiaphragmatic RF
stomach, and gallbladder when treating liver
ablation. tumors, and to nerve roots when bone tumors
are treated with RF. Clearly, the simplest way
to avoid such collateral damage is not to treat
tumors that are less than 1 cm from sensitive
cult or impossible via a transhepatic route as organs. Another option is to refer the patient
well. Transpulmonary access has been de- for a preoperative or laparoscopic ablation pro-
scribed for ethanol injection in hepatocellular cedure during which the untargeted organ will
carcinomas, but pneumothoraces occur in 30% be manually moved away from the targeted
of the cases and require a chest tube in 7% (17). organ. Finally, when a percutaneous approach
Even if some ablative techniques, such as is required, one can try to move the tumor away
radiofrequency (RF), are used in the lung, in from the untargeted organ. A thin needle can
our opinion it is safer to try and avoid collat- be inserted in the narrow or virtual space
eral lung damage. When transpleural access is between the two organs for this purpose, and
needed, we first induce a pneumothorax to then a few milliliters of air or saline (a test
retract the lung toward its hilum. A path for the amount) are injected. Once injection in the
ablation instruments is thus created in a pleural targeted space has been confirmed, a larger
cavity that is transiently devoid of lung paren- amount of air/saline can be used. This technique
chyma and filled with iatrogenic air (Fig. 9.2). can be used, for example, in the peritoneum
The pneumothorax is induced with an epidural in an attempt to widen the space between a
needle by maintaining positive pressure on the peripheral liver tumor and a sensitive organ
piston of an air-filled syringe during the punc- such as the colon or stomach (Fig. 9.3). In this
ture in a similar manner to an epidural injec- setting, CT imaging provides a unique and
tion. When the needle tip reaches the pleural accurate view of any structure, as well as the
space, the decrease in resistance will allow a few injected air or saline.
milliliters of air to be injected. CT is then used Positioning of the ablation instrument in
to check that the needle is positioned correctly relation to the tumor under CT guidance is akin
and that air has been injected in the right to placing the needle in a biopsy procedure.
location. Then, the amount of air needed to However, a major difference between ablation
move the lung away from the future needle and biopsy is that sampling in the latter can be
path is aspirated through a filter, and manually performed practically anywhere in the tumor
9. Computed Tomography Imaging for Tumor Ablation 109

A B

c D
FIGURE 9.3. (A) A CT scan shows a segment I liver the needle, allowing the gastric wall to be moved
metastasis abutting on the gastric wall, which devel- away from the subcapsular metastasis in segment 1.
oped after liver surgery. (B) The tip of a 22-gauge (D) A cool-tip RF needle is placed in the metastasis
Chiba needle, inserted via a transhepatic route under in segment I and thermal ablation can be performed
CT guidance, is located in the narrow space between without collateral damage to the stomach. (Courtesy
the stomach and segment I of the liver. (C) Ten mil- of Dr. Philippe Brunner.)
liliters of air have been manually injected through

provided a necrotic center, if present, is importance. Multiple punctures can lead to


avoided. During ablation therapy, the active tip tumor cell spillage and seeding. This is also per-
of the probe must be positioned in a precise tinent when expandable multiple-array needles
location inside the targeted tumor. Most of the are used for RF ablation; being in the right loca-
time the ablation site should be in the center of tion is critical to avoid repeated deployment of
the tumor so that the entire lesion is destroyed, the hook-shaped inner electrodes, as this is
along with a rim of healthy tissue. Entering the likely to promote tumor seeding.
tumor only once without completely traversing Accurate positioning of the ablation instru-
it with the tip of the needle is of paramount ment in relation to the tumor usually is evalu-
110 T. de Baere

ated on axial images without contrast injection. capsular, it is always preferable to first tra-
In some instances, contrast injection can be verse healthy liver parenchyma to avoid bleed-
useful, particularly when the contrast between ing and tract seeding. Indeed, tumor seeding
the tumor and the surrounding liver is low. Thin along the needle tract has been reported to
control scans should be used, because although occur particularly after treatment of subcap-
thick scans make it easier to visualize the sular hepatocellular carcinomas. Treating the
lesion, they are less accurate to judge the probe tract with cauterization is a potential option to
position in relation to the target. Moreover, it try to minimize such seeding and bleeding. A
has been demonstrated that the use of multi- word of warning: Treatment of the liver capsule
planar reformation in the coronal and sagittal is always painful and thus requires deep
planes statistically improves the accuracy of sedation.
instrument placement. In 44% of cases, instru- Computed tomography guidance is often
ment repositioning was required under imaging used for bone tumors because of the high
guidance in the axial plane (19). quality of visualization it provides. Planning an
When tumors are larger than the volume of entry point perpendicular to the bone surface
ablated tissue that can be obtained in one ap- helps avoid slippage and potential damage to
plication, overlapping ablation is needed to other organs. Drilling the cortical bone is often
attempt to destroy the tumor completely. When required to reach a deep-seated lesion. For
such overlapping ablations are needed, CT example, a safe anatomic entrance is sometimes
probably has an edge over ultrasound for moni- via the normal cortex on the opposite side.
toring. The overriding advantage is that the After drilling, a sheath or a guiding needle can
probe position is easier to visualize with CT be useful to exchange the drill for the ablation
than with US after changes caused by pre- instrument. It is therefore advisable to use a
vious ablations. Indeed, the slight decrease in drill needle that is at least one gauge larger than
Hounsfield units (HU) exhibited by the the ablation tool. If the external metallic sheath
thermal lesions on CT (see Image Monitoring, of the drill system is used to guide the ablative
below) does not impede subsequent instrument tool for RF or microwave ablation, one should
placement at all, and can even be useful for retract the sheath a few centimeters before
guidance. The second advantage is that CT mul- power deposition to avoid contact between the
tiplanar reconstruction can probably help to active part of the probe and the metallic sheath;
position the probes equidistantly in any direc- this contact could cause burns along the track
tion, and that would be ideal placement. In and must be avoided (Fig. 9.4). A plastic sheath
contrast, postablation US imaging demons- is preferred for cryotherapy to avoid freezing
trates either hyperechoic or hypoechoic tissue of the track.
changes, but always with posterior shadowing Osteoid osteoma is a good indication for
that impedes visualization of the tumor or ablation. Laser and RF have both been
needle. Subsequent probe placement is diffi- reported to be effective to treat osteoid osteo-
cult, and inevitably inaccurate, even if the basic mas (22-24). More sophisticated and larger
recommendation to treat the deepest part of caliber devices such as cooled or expandable
the tumor first is followed. Whatever the guid- multiple-array needles are not required for
ance system used, it should be borne in mind osteoid osteomas, as the target is small and a
that if overlapping deliveries are to be per- single bare laser fiber or a single RF needle is
formed, six deliveries will only increase the sufficient. On the other hand, in the case of
diameter of the thermal lesion by 25% (20), and large osteolytic bone tumors, good results
the efficacy of local ablation decreases rapidly have been demonstrated with the expandable
when the size of the target increases (21). multiple-array needle probe for treatment of
Liver tumors should be punctured via the pain in cases refractory to standard manage-
shortest route possible, avoiding large vessels, ment with analgesics and radiation therapy
and, of course, the liver hilum, as well as the (25). When used to manage pain, this particular
gallbladder. However, when tumors are sub- needle is usually chosen to target the margin of
9. Computed Tomography Imaging for Tumor Ablation 111

B
FIGURE 9.4. (A) A CT scan of second lumbar verte- level of the cortical bones, to avoid contact between
bral body lytic metastasis from breast cancer, which the metallic sheath and noninsulated distal part of
has been accessed via a transpedicular approach. A the needle. (B) Two months after RF delivery, CT
single cool-tip RF needle has been inserted through scan shows a rim of hyperattenuating tissue, seem-
the metallic sheath of the drill needle. A few cen- ingly delimiting the external borders of the ablated
timeters of the sheath have been retracted, up to the area.

metastases that involve bones with the objec- mended that only one lung be treated at a time
tive of treating the soft tissue-bone inter- to avoid life-threatening complications that can
face, especially when the entire tumor volume occur in cases of bilateral adverse events such
cannot be destroyed because the metastases are as massive hemorrhage (28) or pneumothorax.
large. It is straightforward to achieve such tar- Notwithstanding, several tumors can be treated
geting under CT guidance, and is important on one side during the same session. Using an
because sensory nerve fibers involving the bone expandable needle for lung RF ablation may be
periosteum and cortex can be destroyed to an advantage, as this type of needle will not slip
inhibit pain transmission. The expandable out of the tumor in case of a massive pneu-
multiple-array RF needle is difficult to deploy mothorax that can push the target tumor away
when dealing with large osteoblastic bone from the needle. The physician should be ready
tumors. Nonexpandable probes are preferred in to insert a chest tube if a significant pneumo-
this situation. thorax occurs. Computed tomography imaging
When treating lung tumors, the needle path is useful for chest tube placement if a postabla-
should traverse the shortest length of aerated tion pneumothorax occurs while the patient is
lung parenchyma, avoiding fissures and mini- still on the CT table. We usually try to manually
mizing the number of passes through the expel the pneumothorax by inserting a 5-
pleural surfaces. Transgression of fissures and French side-hole catheter in the air-filled
pleural surfaces increases the incidences of pleural cavity under CT guidance. This catheter
pneumothoraces (26). Furthermore, trying to is connected to a 50-mL syringe via a three-way
insert a needle as perpendicular as possible to stopcock; air is drawn from the pneumothorax
the pleura also appears to be important in into the syringe and then expelled. Larger
averting the risk of pneumothorax (27). What- caliber chest tubes are used only in cases of
ever the ablation technique used, it is recom- recurrent pneumothorax.
112 T. de Baere

Image Monitoring for the measurement of ablated tissue. Ultra-


sound slightly underestimated the true size of
As no imaging technique is capable of confirm- ablated tissue. Better correlation with the
ing cell death with 100% accuracy, there is no pathologic study also has been reported for CT
way of knowing whether the tumor has been monitoring versus intraoperative US in the
completely destroyed at the time of treatment. kidney (32).
Image monitoring of the ablation procedure As enhanced CT provides a more clear-cut
rarely examines the extent of treatment delineation of the ablated area than unen-
directly. Only MR thermometry directly evalu- hanced CT, enhanced CT is performed at the
ates the extent of heat or cold. All the other end of the treatment to try to best determine
imaging techniques simply image changes that the extent of coagulation. The goal of ablation
are indirect consequences of treatment. These is to obtain an unenhanced area that is at least
modifications do not correspond exactly to the as large as the tumor, and ideally a few mil-
extent of the ablated volume, nor to certain limeters larger. Of course, if the targeted tumor
cell death within the volume. In the liver, for exhibits high enhancement before ablation,
example, US monitoring depicts a hyperechoic disappearance of this enhancement must be
area induced by the formation of gas after RF, achieved. If tumor enhancement persists, CT
laser, or microwave ablation; US demonstrates allows retargeting of residual foci during the
a hypoechoic iceball after cryotherapy, and same treatment session. As the amount of con-
hyperechoic changes after alcohol injection. trast medium that can be injected is limited
The consequences of treatment depicted by and enhancement is less intense after subse-
MR imaging are areas of hypointense signal quent injections, enhanced imaging cannot be
following cryotherapy and RF, and/or devascu- repeated at will. The use of contrast medium at
larized tissue that any ablative technique can CT optimally is left for the final evaluation.
produce. During chemical ablation, CT is able to
Most of the data and experience with CT image the spatial diffusion of the injected
monitoring of tissue ablation are related to RF liquid. Alcohol is slightly hypoattenuating, but
ablation. RF ablation in the liver was well any toxic liquid or gel can be transformed into
described in an animal study by Cha et al (29), a hyperattenuating substance by adding con-
in which CT was performed immediately and 2 trast medium to the therapeutic compound, as
and 8 minutes after treatment. Unenhanced CT reported for alcohol (33,34) or chemothera-
scans 2 minutes after RF showed a 14-HU peutic drugs (35). Once again, although imaging
decrease in ablated tissue compared to healthy is capable of depicting the diffusion of the toxic
liver, and this difference increased to 22HU at agent, it cannot predict the efficacy of the treat-
8 minutes. After cryotherapy, frozen tissue also ment. Here again, injection of contrast medium
was described as a well-demarcated region of can help to evaluate the extent of coagulation
lower attenuation than normal liver (30). necrosis by imaging the unenhanced area; con-
After hyperthermia or cryoablation, the versely, an area of the tumor with persistent
postablation hypoattenuating area on CT often enhancement can be subsequently targeted for
contains several small gas bubbles of various retreatment, as reported for alcohol therapy of
sizes that are irregularly distributed in the hepatocellular carcinomas (HCCs) (4).
ablated tissue. After injection of contrast Computed tomography monitoring of ther-
medium, the differences between RF-ablated mal ablation in bone or kidney tumors delin-
tissue and healthy liver increase to 55 HU, with eates roughly the same features as described
improved conspicuity and edge detection. CT previously in the liver. The formation of gas
was found to correlate better than US with the bubbles at irregularly distributed points within
real volume of ablated liver tissue measured at the ablated lesion is common. If the tumor is
pathologic examination (29,31); the peripheral initially highly enhanced, this enhancement
rim of enhancement that corresponds to an disappears if treatment is efficient; otherwise
inflammatory reaction is not taken into account there is a small decrease in density.
9. Computed Tomography Imaging for Tumor Ablation 113

In the lung, RF ablation causes lung tissue to treatment. It becomes more intense and slightly
became more dense in the ablated area more extensive during the following hours and
(Fig. 9.5). This is akin to alveolar consolidation, days (see Follow-Up Imaging, below). Although
and is often very faint during the course of it is unclear whether these CT imaging changes

A B

c D

FIGURE 9.5. (A) Prone CT scan of the lungs shows an


expandable RF needle inserted in a 17-mm lung metas-
tasis from colon cancer. Note the small pneumothorax,
which will not necessitate any aspiration or drainage. (B)
Lung tissue consolidation around the treated tumor can
be seen after 17 minutes of RF ablation. (C) One day
later, the post-RF alveolar condensation has increased
in size and density. There is a little pleural effusion.
(D) Two months later, there is retraction and increased
density of the thermally destroyed area. (E) Four
months after treatment, the scar is even smaller. E
114 T. de Baere

really represent coagulation necrosis, this area


of density is helpful when attempts are made to
shape the thermal lesion so that it is equivalent
to the tumor size.

Follow-Up Imaging
As stated earlier, there is no way of affirming
that a tumor has been completely ablated during
the procedure itself. Consequently, follow-up
imaging is crucial to determine treatment effi-
ciency, or, on the contrary, local failure of abla-
tion. When failure occurs, follow-up imaging is
used to image the location of residual tumor
accurately for further ablative therapy. As the
tumor size is a critical factor in the efficacy FIGURE 9.6. Four months after RF ablation of a
of ablation, it is important that regrowth be metastasis from colon cancer, CT scan shows a
unveiled as early as possible to optimize re- hypoattenuating unenhanced round area corre-
treatment of small lesions. Thus, follow-up sponding to ablated tissue. The heterogeneously
imaging should be performed at regular enhanced curviform area at the anteromedial border
of the scar, in contact with the middle hepatic vein,
intervals and over a long period of time, be-
has appeared since the previous CT scan performed
cause incomplete ablation often leaves small 2 months earlier. This is tumor regrowth from resid-
undetectable tumor foci that will be visible on ual unablated foci. This incomplete ablation has
delayed imaging only when the residual cells probably been promoted by heat sink induced by the
have grown to form a larger tumor (Fig. 9.6). neighboring large hepatic vein.
As the aim of ablation is to generate an area
of necrosis, the diameter of which is larger or at
least equivalent to that of the tumor. Ideally, the
destroyed tissue or "scar" should be larger than mean of 45% at 6 months and 63% at 12
the treated lesion, encompassing the tumor and months (37).
safety margins. If treatment is successful, this World Health Organization (WHO) criteria
scar will decrease in size, albeit slowly. Indeed, or Response Evaluation Criteria in Solid
in our experience with mostly colon cancer Tumors (RECIST) criteria (38,39) cannot be
metastases in the liver, 66% of successfully applied to assess response to ablative therapies,
treated tumors decreased in size during 12 at least not early during the posttreatment
months of imaging follow-up. The decrease in period. By these criteria, anticancer treatment
the product of the two largest dimensions of the is based on a decrease in tumor size, which
scar attained a mean of 15% (range 10-30%) is not really applicable to ablation. Other
at 6 months, and 35% (range 15-90%) at 12 methods are needed to assess the efficacy of
months (36). A series of 43 hepatocellular car- ablative treatment. Today, there is no clear con-
cinomas treated with RF showed a decrease sensus about which imaging techniques are the
in the immediate postablation scar volume of most appropriate for follow-up after ablation
21 % at 1 month, 50% at 4 months, 65% at 7 and various imaging techniques are used from
months, and 89% at 16 months. This slow one study to another. However, the criteria
decrease in the scar volume has also been most commonly used to assess this efficacy on
reported after cryotherapy (30). Ethanol- CT, MR, or US imaging is the absence of tissue
induced necrosis seems to decrease in size more enhancement, probably while awaiting func-
often and faster, with reports of a diminution tional imaging (positron emission tomography
of size in all treated tumors that attained a [PET] scanning).
9. Computed Tomography Imaging for Tumor Ablation 115

Liver
Enhanced CT and MR imaging are at present
considered the most useful modalities to assess
thermal ablation efficacy in the liver. Prelimi-
nary reports suggest that the size of the nonen-
hancing region after RF visualized at CT and
MR imaging corresponded to within 2 mm of
the size of the necrosis measured histologically
(40-42) or with cryotherapy (43).
When RF therapy is successful, a hypoatten-
uating well-demarcated, round or oval scar is
depicted on CT. Areas of higher attenuation
can be found within the thermally ablated area
after RF, microwave, laser, or cryotherapy, FIGURE 9.8. A CT scan 2 months after RF ablation
corresponding to hemorrhage. The thermally of a subcaspsular metastasis shows a thin peripheral
ablated area does not enhance after injection of rim of enhancement corresponding to inflammatory
contrast medium. tissue.
Two particular postthermoablation imaging
patterns must be underscored. The first is the
presence of wedge-shaped areas of contrast
enhancement abutting on the RF scar seen in contrast enhancement are probably due to per-
12% of patients in the arterial phase on CT fusion disorders, such as small-vessel throm-
(Fig. 9.7) (36). These wedge-shaped areas of bosis and arterioportal fistulas induced by
ablation. The second pattern found after any
type of ablation is a thin peripheral rim of
enhancement, usually measuring less than 2mm
in width, which is often seen surrounding the
radiofrequency scars (Fig. 9.8). This rim is due
to inflammatory and granulation tissue with
profuse neovascularization found after most
ablative techniques, as that described in an
experimental model after focused US (44) or
cryotherapy (43), as well as in clinical practice
after RF (36,40,45). This rim has been described
less commonly in hepatocellular carcinoma
treated with alcohol injection (46,47) and in
hepatic metastases treated with laser-induced
thermotherapy (48,49). The inflammatory rim
decreases with time. We found this rim in 24%
and 18% of RF-treated tumors, respectively, on
MR and CT at 2 months, in 4% on MR at 4
months, and in 2% on MR at 9 months (36).
FIGURE 9.7. Two months after RF ablation of a
Others have reported this rim in 89% of CT
breast cancer metastasis, the scar is not enhanced.
studies after RF treatment within 1 month, in
There is a large wedge-shaped perfusion disorder
demonstrating early and heavy enhancement of 56% between 1 and 3 months, and in 22%
the parenchyma in segment VII. Note the faint between 3 to 6 months (45).
dilation of the biliary tract at the periphery of the Residual active tumor foci depicted on CT
scar, imaged as periportal hypoattenuatting linear usually are found at the periphery of the RF-
structures. induced lesion, and can appear as a nodule or
116 T. de Baere

local thickening of the peripheral rim. More and MR found eight. However, equivalent
rarely, an increase in the size of the treated area imaging results were obtained at 4 months with
signifies incomplete treatment. This increase in both imaging techniques, which revealed eight
size, also called the halo pattern of recurrence, of nine incomplete treatments, with the ninth
is found more frequently in recurrent metas- being discovered at 6 months. TI-weighted
tases than with HCC (50). images were the most reliable sequences, due to
Foci of active tumor exhibit hypo- or hyper- good contrast between the hypointense scar
attenuation on CT, and HCC's usually demon- and the mildly hyperintense residual tumor.
strate contrast enhancement during the arterial However, caution should be exercised when
phase. In contrast, metastases remain mostly distinguishing the mildly hyperintense signal
hypovascular, and sometimes demonstrate indicative of residual tumor from the heavily
low-grade enhancement at their periphery on hyperintense, fluid-like appearance of liquid
imaging of the portal phase. The arterial phase necrosis.
image is therefore best to detect recurrent Contrast-enhanced harmonic power Doppler
HCC, while the portal phase image is best to US, compared to CT, appears to be useful to
depict recurrent colorectal metastases. Due to demonstrate local relapse after RF treatment.
the heat sink effect encountered during thermal The technique demonstrated a sensitivity of
ablation with RF, considerable attention should 90% to 98%, a specificity of 100%, and 98%
be paid to scars located in the vicinity of large accuracy, in detecting local recurrence 3 to
vessels where incomplete treatment is seen 7 days after RF ablation of hypervascular
more frequently (42,51). HCC's (54,55). However, this technique is
A major question is when postablation much less efficient in detecting residual tumor
imaging follow-up should be initiated and how after treatment of colorectal metastases.
regularly it should be repeated. It is difficult to Among 75% of tumors that showed enhance-
appraise results soon after treatment because ment before treatment, only 50% of the post-
of the peripheral inflammatory rim that RF local relapses were seen the day after
enhances and can be misinterpreted as residual treatment (52).
tumor, or small foci of remnant tumor embed- Imaging of complications is an important
ded in this rim that may be overlooked. These reason for follow-up imaging, and CT has a
are reasons why the first imaging studies are major role to play in this context. One of the
usually performed 1 to 3 months after RF, and more common complications after ablation
then repeated every 2 or 3 months. Repeat therapy is an abscess in the liver after RF
imaging eventually will demonstrate regrowth (56,57), and to a lesser extent after cryotherapy
of small foci of unablated tumor that remain in (58). This complication seems to be much more
the scar, but were too small to be detected frequent in patients who have a bilioenteric
earlier. However, for most authors, including anastomosis. These abscesses typically have
Solbiati et al (52), who reported 77% occurring been reported after an interval that ranges from
before 6 months, and 96% depicted by 1 year, 8 days to 5 months (51,56,57). Clinical symp-
the likelihood of such local regrowth after 12 toms may be mild, so imaging plays a major role
months is low (36,53). This signifies that after 1 in their depiction. They usually appear as a gas-
year, the main aim of follow-up imaging is to containing mass that has developed in the
search for new hepatic tumors rather than local ablated bed. They usually can be drained under
recurrences. CT guidance. However, small gas bubbles are
In most studies, CT and MR are used vari- nearly always imaged during ablation either
ably to assess the efficacy of RF treatment. We with hyperthermia or cryotherapy, and they can
compared CT and MR in our early experience persist for several days. In our experience with
in RF of liver tumors, imaging every 2 months RF ablation, all the ablated areas containing
with both CT and MR (36). Incomplete treat- gas after 2 weeks always were aspiration-
ment was depicted in nine cases. At 2 months proven abscesses. However, gas bubbles have
of follow-up, CT revealed four local failures been described in the liver several weeks after
9. Computed Tomography Imaging for Tumor Ablation 117

cryotherapy without a related infection (30). Bones


Biliary tract dilation upstream from an ablated
area is common, and found in 8% of follow-up Computed tomography imaging after ablation
imaging in our hands (Fig. 9.7). This condition of bone tumors demonstrates late changes.
was always clinically asymptomatic in our expe- After ablation of an osteoid osteoma, healing
rience, as no tumors were targeted for ablation of the ablated area usually takes more than 1
close to the liver hilum. year. As a small volume is treated, usually with
a bare laser fiber or a single RF needle, no
changes are usually found in the bone sur-
rounding the ablated lesion; however, early
Lung pain relief (usually in less than a week), is the
One or 2 days after RF ablation, CT imaging best sign that therapy has been effective.
depicts an increase in the size and density of the In the case of malignant bone tumors, late
blurred parenchyma surrounding the treated signs of efficacy are a shrunken ablated lesion
tumor, compared to that imaged immediately and healing of bone destruction. No enhance-
after the ablation procedure (Fig. 9.5). This ment in ablated areas signifies the absence of
increase in the size of the ablated lesion varies viable tumor, as in other organs. Furthermore,
considerably among patients. It can be up to 5 after a few weeks, CT often shows a rim of
to 10mm and usually is concentric, but can hyperattenuating bone tissue that outlines the
cover a complete lung segment with a blurred external borders of the ablated area (Fig. 9.4),
outline, unless the border is sharply defined by although the true explanation for this appear-
the neighboring fissure. A small pleural effusion ance remains obscure. Spontaneous high bone
is nearly always seen during the early post- density prohibits the study of rim contrast
treatment period. After 1 to 2 months, this large enhancement with CT. However, on MR, this
and blurred area of consolidation shrinks, and rim appears as low signal on Tl-weighted
forms a scar with a sharper border and higher images, and high signal on T2-weighted images.
tissue density. This scar usually continues to be Enhanced signal occurs after injection of
slightly larger than the targeted tumor. If the gadolinium. These imaging patterns are similar
treatment is successful, the scar will continue to to those of the inflammatory rim described in
shrink, but slower than during the first 2 the liver after all types of ablative therapy, and
months, akin to that reported for liver (Fig. 9.5). therefore confirm the hypothesis that this is the
Using contrast-enhanced CT to assess the external border of the ablated area.
efficacy of lung RF ablation has yet to be eval-
uated, but probably depends on the type of
enhancement of the tumor before treatment.
Kidney
Late complications are rare. Early and midterm The size and enhancement criteria used to
complications are mostly pleural effusions and assess treatment efficacy in the liver with CT
pneumothoraces. Hemorrhage is unusual in the are also valid for kidney tumors after ablation.
lung parenchyma even though minor hemopty- However, stranding of the perirenal fat, usually
sis is common from 1 to 10 days after the pro- roughly parallel to the ablated area is an addi-
cedure. One case of major hemorrhage has tional feature that evolves on sequential follow-
been reported (28), and avoiding major vessels up CT scans into a more organized rim of
during the puncture may be a way to avert such hyperattenuating tissue, seemingly delineating
complications. Finally, sepsis may appear as seg- the external borders of the ablated area (Fig.
mental or lobar consolidation on imaging that 9.9) (59,60). Wedge-shaped nonenhancing areas
represents a pneumonia. In cases oflong-Iasting in the kidney adjacent to the ablated lesion are
fever, CT should be used to search for an seen on more than half of the follow-up CT
abscess at the ablation site that generally images performed at 2 months. They are prob-
appears as a partially air-filled cavity instead of ably due to peripheral renal infarcts after co-
the usual scar tissue. agulation of small arterial feeding branches. A
118 T. de Baere

6. Paulson EK, Sheafor DH, Enterline DS, et al. CT


fluoroscopy-guided interventional procedures:
techniques and radiation dose to radiologists.
Radiology 2001;220:161-167.
7. Yueh N, Halvorsen RA Jr, Letourneau JG, Crass
JR. Gantry tilt technique for CT-guided biopsy
and drainage. J Comput Assist Tomogr 1989;13:
182-184.
8. Kirchner J, Kickuth R, Walz MV, et al. CTF-
guided puncture of an unenhanced isodense
liver lesion during continuous intravenous injec-
tion of contrast medium. Cardiovasc Intervent
Radiol 1999;22:528-530.
9. Schweiger GD, Brown BP, Pelsang RE, Dhadha
RS, Barloon TJ, Wang G. CT fluoroscopy: tech-
nique and utility in guiding biopsies of tran-
FIGURE 9.9. Follow-up CT scan obtained 8 months siently enhancing hepatic masses. Abdom Imag-
after RF ablation of an exophytic renal carcinoma. ing 2000;25:81-85.
The unenhanced scar is composed of ablated tumor 10. Jacobi V, Thalhammer A, Kirchner 1. Value of a
and a few millimeters of enhanced renal parenchyma laser guidance system for CT interventions: a
adjacent to its inner limit. There is a thin rim of hyper- phantom study. Eur RadioI1999;9:137-140.
attenuating tissue in the perirenaVperitumorous fat, 11. Holzknecht N, HeImberger T, Schoepf UJ, et al.
a few millimeters from the ablated area. [Evaluation of an electromagnetic virtual target
system (CT-guide) for CT-guided interventions].
Rofo Fortschr Geb Rontgenstr Neuen Bildgeb
small amount of peri- or pararenal blood is seen Verfahr 2001;173:612--618.
12. Pereles FS, Baker M, Baldwin R, Krupinski E,
in 10% to 40% of ablated renal lesions, but it
Unger EC. Accuracy of CT biopsy: laser guid-
always disappears on the first follow-up CT at ance versus conventional freehand techniques.
1 or 2 months (59,60). Acad RadioI1998;5:766-770.
13. Goodacre BW, Savage C, Zwischenberger JB,
References Wittich GR, vanSonnenberg E. Salinoma window
technique for mediastinal lymph node biopsy.
1. Sheafor DH, Paulson EK, Kliewer MA, DeLong Ann Thorac Surg 2002;74:276-277.
DM, Nelson RC. Comparison of sonographic 14. Langen HJ, Klose KC, Keulers P, Adam G,
and CT guidance techniques: does CT fluoros- Jochims M, Gunther RW. Artificial widening of
copy decrease procedure time? AJR 2000;174: the mediastinum to gain access for extrapleural
939-942. biopsy: clinical results. Radiology 1995;196:703-
2. Kirchner J, Kickuth R, Laufer U, Schilling EM, 706.
Adams S, Liermann D. CT fluoroscopy-assisted 15. Karampekios S, Hatjidakis AA, Drositis J, et al.
puncture of thoracic and abdominal masses: a Artificial paravertebral widening for percuta-
randomized trial. Clin RadioI2002;57:188-192. neous CT-guided adrenal biopsy. J Comput
3. Teeuwisse WM, Geleijns J, Broerse JJ, Assist Tomogr 1998;22:308-310.
Obermann WR, van Persijn van Meerten EL. 16. Shah H, Harris VJ, Konig CW, et al. Saline injec-
Patient and staff dose during CT guided biopsy, tion into the perirectal space to assist trans-
drainage and coagulation. Br J Radiol 2001;74: gluteal drainage of deep pelvic abscesses. J Vasc
720-726. Interv RadioI1997;8:119-121.
4. Takayasu K, Muramatsu Y, Asai S, Kobayashi T. 17. Shibata T, Iimuro Y, Yamamoto Y, et al. CT-
CT fluoroscopy-assisted needle puncture and guided transthoracic percutaneous ethanol injec-
ethanol injection for hepatocellular carcinoma: a tion for hepatocellular carcinoma not detectable
preliminary study. AJR 1999;173:1219-1224. with US. Radiology 2002;223:115-120.
5. Kato R, Katada K, Anno H, Suzuki S, Ida Y, 18. Elias D, de Baere T, Goharin A, Lasser P, Roche
Koga S. Radiation dosimetry at CT fluoroscopy: A. Transpleurodiaphragmatic radiofrequency
physician's hand dose and development of thermoablation of a liver metastasis. J Am Coil
needle holders. Radiology 1996;201:576-578. Surg 2000;191:683-685.
9. Computed Tomography Imaging for Tumor Ablation 119

19. Antoch G, Kuehl H, Vogt F, Debatin J, Stattaus computed tomography-guided percutaneous


1. Value of CT volume imaging for optimal place- radiofrequency ablation of renal tissue: acute
ment of radiofrequency ablation probes in liver and chronic effects in an animal model. Urology
lesions. J Vasc Interv RadioI2002;13:1155-1161. 2001;57:976-980.
20. Dodd GD III, Frank MS,Aribandi M, Chopra S, 33. Hamuro M, Kaminou T, Nakamura K, et al.
Chintapalli KN. Radiofrequency thermal abla- Percutaneous ethanol injection under CT
tion: computer analysis of the size of the thermal fluoroscopy for hypervascular hepatocellular
injury created by overlapping ablations. AJR carcinoma following transcatheter arterial
2001;177:777-782. embolization. Hepatogastroenterology 2002;49:
21. Livraghi T, Goldberg SN, Lazzaroni S, et al. 752-757.
Hepatocellular carcinoma: radio-frequency 34. Ueda K, Ohkawara T, Minami M, et al. [Non-
ablation of medium and large lesions. Radiology real-time computed tomography-guided per-
2000;214:761-768. cutaneous ethanol injection therapy for
22. Gangi A, Dietemann JL, Gasser B, et al. Inter- hepatocellular carcinoma undetectable by ultra-
stitial laser photocoagulation of osteoid osteo- sonography]. Gan To Kagaku Ryoho 1998;25:
mas with use of CT guidance. Radiology 1997; 1254-1258.
203:843-848. 35. Farres M, de Baere T, Lagrange C, et al. Percu-
23. Rosenthal DI. Percutaneous radiofrequency taneous mitoxantrone injection for primary and
treatment of osteoid osteomas. Semin Muscu- secondary liver tumors: preliminary results. Car-
loskelet RadioI1997;1:265-272. diovasc Intervent Radiol 1998;21:399-403.
24. Rosenthal DI, Hornicek FJ, Wolfe MW, Jennings 36. Dromain C, de Baere T, Elias D, et al. Hepatic
LC, Gebhardt MC, Mankin H1. Percutaneous tumors treated with percutaneous radiofre-
radiofrequency coagulation of osteoid osteoma quency ablation: CT and MR imaging follow-up.
compared with operative treatment. J Bone Joint Radiology 2002;223:255-262.
Surg [Am] 1998;80:815-821. 37. Ebara M, Kita K, Sigiura YM, et al. Therapeutic
25. Callstrom MR, Charboneau JW, Goetz MP, effect of percutaneous ethanol injection on small
et al. Painful metastases involving bone: feasi- hepatocellular carcinoma: evaluation with CT.
bility of percutaneous CT- and US-guided radio- Radiology 1995;195:371-377.
frequency ablation. Radiology 2002;224:87-97. 38. Miller AB, Hoogstraten B, Staquet M, Winkler
26. Haramati LB, Aviram G. What constitutes effec- A. Reporting results of cancer treatment. Cancer
tive management of pneumothorax after CT- 1981;47:207-214.
guided needle biopsy of the lung? Chest 2002; 39. Padhani AR, Ollivier L, Shibata T, et al. The
121:1013-1015. RECIST (Response Evaluation Criteria in Solid
27. Saji H, Nakamura H, Tsuchida T, et al. The inci- Tumors) criteria: implications for diagnostic
dence and the risk of pneumothorax and chest radiologists. Br J Radiol 2001;74:983-986.
tube placement after percutaneous CT-guided 40. Goldberg SN, Gazelle GS, Compton CC, Mueller
lung biopsy: the angle of the needle trajectory is PR, Tanabe KK. Treatment of intrahepatic
a novel predictor. Chest 2002;121:1521-1526. malignancy with radiofrequency ablation: radio-
28. Vaughn C, Mychaskiw G, II, Sewell P, et al. logic-pathologic correlation. Cancer 2000;88:
Massive hemorrhage during radiofrequency 2452-2463.
ablation of a pulmonary neoplasm. Anesth 41. Morimoto M, Sugimori K, Shirato K, et al.
Analg 2002;94:1149-1151. Treatment of hepatocellular carcinoma with
29. Cha CH, Lee Ff Jr, Gurney JM, et al. CT versus radiofrequency ablation: radiologic-histologic
sonography for monitoring radiofrequency abla- correlation during follow-up periods. Hepatology
tion in a porcine liver. AJR 2000;175:705-711. 2002;35:1467-1475.
30. McLoughlin RF, Saliken JF, McKinnon G, 42. Solbiati L, Livraghi T, Goldberg SN, et al. Per-
Wiseman D, Temple W. CT of the liver after cutaneous radio-frequency ablation of hepatic
cryotherapy of hepatic metastases: imaging find- metastases from colorectal cancer: long-term
ings. AJR 1995;165:329-332. results in 117 patients. Radiology 2001 ;221:
31. Raman SS, Lu DS,Vodopich DJ, Sayre J, Lassman 159-166.
C. Creation of radiofrequency lesions in a 43. Kuszyk BS, Boitnott JK, Choti MA, et al. Local
porcine model: correlation with sonography, CT, tumor recurrence following hepatic cryoa-
and histopathology. AJR 2000;175:1253-1258. blation: radiologic-histopathologic correlation
32. Crowley JD, Shelton J, Iverson AJ, Burton Mp, in a rabbit model. Radiology 2000;217:477-
Dalrymple NC, Bishoff JT. Laparoscopic and 486.
120 T. de Baere

44. Rowland I, Rivens I, Chen L, et al. MRI study of ment with US microbubble contrast agent-
hepatic tumours following high intensity focused early experience. Radiology 1999;211:643-649.
ultrasound. Br J RadioI1997;70:144-153. 53. Curley SA, Izzo F, Ellis LM, Vauthey IN, Vallone
45. Tsuda M, Majima K, Yamada T, Saitou H, P. Radiofrequency ablation of hepatocellular
Ishibashi T, Takahashi S. Hepatocellular carci- cancer in 110 patients with cirrhosis. Ann Surg
noma after radiofrequency ablation therapy: 2000;232:381-391.
dynamic CT evaluation of treatment. Clin 54. Cioni D, Lencioni R, Rossi S, et al. Radiofre-
Imaging 2001;25:409-415. quency thermal ablation of hepatocellular carci-
46. Ito K, Honjo K, Fujita T, Awaya H, Matsumoto T, noma: using contrast-enhanced harmonic power
Matsunaga N. Enhanced MR imaging of the liver Doppler sonography to assess treatment out-
after ethanol treatment of hepatocellular car- come.AJR 2001;177:783-788.
cinoma: evaluation of areas of hyperperfusion 55. Meloni MF, Goldberg SN, Livraghi T, et al.
adjacent to the tumor.AJR 1995;164:1413-1417. Hepatocellular carcinoma treated with radiofre-
47. Sironi S, De Cobelli F, Livraghi T, et al. Small quency ablation: comparison of pulse inversion
hepatocellular carcinoma treated with per- contrast-enhanced harmonic sonography,
cutaneous ethanol injection: unenhanced and contrast-enhanced power Doppler sonography,
gadolinium-enhanced MR imaging follow-up. and helical CT. AJR 2001;177:375-380.
Radiology 1994;192:407-412. 56. Wood TF, Rose DM, Chung M, Allegra DP,
48. Amin Z, Donald JJ, Masters A, et al. Hepatic Foshag U, Bilchik AJ. Radiofrequency ablation
metastases: interstitial laser photocoagulation of 231 unresectable hepatic tumors: indications,
with real-time US monitoring and dynamic eval- limitations, and complications. Ann Surg Oncol
uation of treatment. Radiology 1993;187:339- 2000;7:593-600.
347. 57. Zagoria RJ, Chen MY, Shen P, Levine EA. Com-
49. Vogi T, Muller P, Hammersting R, et al. Malig- plications from radiofrequency ablation of liver
nant liver tumors treated with MR imaging- metastases. Am Surg 2002;68:204-209.
guided laser-induced technique: technique and 58. Bilchik AJ, Wood TF, Allegra D, et al. Cryosurgi-
prospective results. Radiology 1995;196:257-265. cal ablation and radiofrequency ablation for
50. Chopra S, Dodd GD III, Chintapalli KN, unresectable hepatic malignant neoplasms: a pro-
Leyendecker JR, Karahan 01, Rhim H. Tumor posed algorithm. Arch Surg 2000;135:657-662;
recurrence after radiofrequency thermal abla- discussion 662-664.
tion of hepatic tumors: spectrum of findings on 59. de Baere T, Kuoch V, Smayra T, et al. Radio-
dual-phase contrast-enhanced CT. AJR 2001; frequency ablation of renal cell carcinoma:
177:381-387. preliminary experience. J Urol 2002;167:1961-
51. de Baere T, Elias D, Dromain C, et al. Radio- 1964.
frequency ablation of 100 hepatic metastases 60. Gervais D, O'Neill M,Arellano R, McGovern R,
with a mean follow-up of more than 1 year. AJR McDougal W, Mueller P. Peritumoral CT
2000;175:1619-1625. changes associated with radiofrequency ablation
52. Solbiati L, Goldberg S, Ierace T, Dellanoce M, of focal renal lesions: description, incidence,
Livraghi T, Gazelle S. Radio-frequency ablation and significance (abst.). Radiology 2OO1;221(P):
of hepatic metastases: post procedural assess- 180.
10
Positron Emission Tomography
Imaging for Tumor Ablation
Annick D. Van den Abbeele, David A. Israel, Stanislav Lechpammer, and
Ramsey D. Badawi

Nuclear Oncology and now found widespread applications in the diag-


nosis, staging, and restaging of these and other
FDG-PET malignancies and has been shown to be as
reliable and accurate as conventional imaging
Nuclear medicine imaging involves the injec- modalities including CT and MRI, or even
tion or ingestion of radioactive pharmaceuticals moreso (6,18-40). Owing substantially to PET
known as radiotracers, each designed to track scanning, nuclear oncology has become an inte-
a particular physiologic or pathophysiologic gral part of the multidisciplinary clinical man-
process. In contrast to conventional radiologic agement of patients with cancer in many
imaging such as x-ray computed tomography institutions (41). Today, nuclear oncology is one
(CT) and magnetic resonance imaging (MRI), of the most dynamic and rapidly growing areas
which map out anatomic structure and depend of contemporary nuclear medicine.
on changes in morphology or size for determi- The evolving roles of noninvasive PET
nation of pathology, nuclear medicine imaging imaging reported in the monitoring of response
provides information on the metabolic function to therapy, and as a predictor of response to
of the investigated organ or tissue. therapy and survival (21,42-62) place FDG-
Fluorine-18-fl uoro-2-deoxy- D-gi ucose PET in an optimal position to evaluate
(FDG) is a glucose analog labeled with the response to tumor ablation procedures.
positron emitter fluorine 18. Positron emission
tomography (PET) with FDG (FDG-PET) is a
whole-body, functional imaging technique that Principles of PET Imaging
measures glucose metabolism. Malignancies
can be detected in resulting images due to their The radionuclides used for the creation of
higher rates of glucose transport compared to labeled radiotracers for PET imaging are
normal tissue (1-4). Most common types of positron emitters, as opposed to the single
malignancies, including cancers of the lung, photon emitters used in conventional nuclear
breast, esophagus, colon, head and neck, and medicine imaging. Positrons do not penetrate
thyroid, as well as lymphomas and melanomas human tissue very far (0-2mm) before com-
show increased FDG uptake (1,4-16). bining with electrons and undergoing annihila-
The Centers for Medicaid and Medicare tion reactions. In such a reaction, a positron and
Services (CMS) have recognized the utility of an electron are converted into two 511-keV
FDG-PET in the management of patients with annihilation photons, emitted at very nearly 180
cancer and have approved reimbursements in degrees to each other. PET cameras usually
all of the aforementioned indications (17). consist of rings of detectors that surround the
Positron emission tomography scanning has patient. When two annihilation photons are

121
122 A.D. Van den Abbeele et al

detected within a certain (-6-12 ns) time frame often have enhanced glucose transport at the
by detectors in these rings, a radioactive decay cell surface, more intense hexokinase activity,
is deemed to have occurred along the line and reduced glucose-6-phosphatase activity.
joining the two detectors involved. In this way,
information regarding the location of radioac-
tivity within the patient can be obtained. Patient Preparation and Normal
Reconstruction of this information yields tomo- Biodistribution of FDG
graphic images estimating the radiotracer con-
centration within the patient. Patients are asked to fast for 4 to 6 hours prior
The radionuclides used for clinical PET to the injection of FDG to maintain low serum
imaging possess relatively short half-lives glucose and endogenous insulin levels. Patients
(approximately between 2 minutes and 2 are also asked to refrain from exercise 24 hours
hours), and most are produced by a cyclotron. prior to the study to minimize striated muscle
The most widely used radionuclide in clinical uptake. A normoglycemic status aids in mini-
PET applications is fluorine 18, which has a mizing competition between circulating glucose
half-life of 109.8 minutes. This half-life is suffi- and FDG at the cellular level, which maximizes
cient to allow outsourcing of radiotracer pro- tumor-to-background ratio and results in
duction to off-site commercial manufacturing optimal lesion detection. However, compliance
facilities. with these prescanning instructions may be
Modern dedicated whole-body PET scanners challenging in diabetic patients, particularly
generate images with a spatial resolution on the those who are insulin-dependent and whose
order of 7 mm, and lesions of that size and disease is poorly controlled. If a patient is dia-
smaller demonstrate a diminished density of betic and insulin-dependent, the injection of
counts compared to larger lesions having FDG is performed approximately 4 hours after
the same biologic characteristics. Lesions below the last insulin injection. A hyperinsulinemic
7 mm in size, therefore, may not be detectable. state favors preferential uptake of FDG by stri-
If the imaging is done using a dual-headed ated muscles and increases the likelihood of
nuclear medicine imaging camera, the smallest a false-negative study. Patients with normal
detectable lesion will be even larger (around blood glucose control also may have a subopti-
15mm). mal study if they do not fast prior to the scan.
Intense tracer uptake in muscle and fat is
sometimes seen in the neck, paraspinal, pec-
FDG-PET Pharmacokinetics toral, and shoulder regions, particularly in
young patients. This uptake may be arneliorated
FDG is transported into metabolically active or reduced with the oral administration of ben-
cells by normal glucose transporter mecha- zodiazepine anxiolytics prior to the injection of
nisms. Once in the cell, FDG is phosphorylated the tracer.
by hexokinase to FDG-6-phosphate in the The brain cortex, basal ganglia, and thalami
same way that glucose is phosphorylated to typically show high FDG uptake. Myocardial
glucose-6-phosphate, but as opposed to uptake is variable, and the fasting state assists
glucose-6-phosphate, FDG-6-phosphate does in reducing the myocardial uptake of FDG.
not undergo further metabolism within the cell. Bowel uptake is also variable, but bowel prepa-
Since FDG-6-phosphate cannot easily cross the rations typically are not used in routine clinical
cell membrane, and since the dephosphoryla- practice since the procedure is similar to that
tion by glucose-6-phosphatase is a relatively used for colonoscopy preparation and may be
slow process in comparison to that of glucose- too demanding for oncologic patients. Low to
6-phosphate, FDG-6-phosphate remains effec- mild diffuse uptake of FDG is usually seen
tively trapped within the cell. The resulting throughout the liver and splenic parenchyma.
buildup of radiotracer within viable tissue is Focal uptake of higher intensity than the
further potentiated in malignant cells, which surrounding parenchyma should raise the
10. Positron Emission Tomography Imaging for Tumor Ablation 123

suspicion for the presence of disease. Of note, Several methods are available for evaluating
attenuation correction artifacts also may mimic therapeutic response using FDG-PET (79-81).
lesions, particularly in the liver. Analysis of The simplest method is visual assessment of
the non-attenuated-corrected images will help tumor uptake by comparing FDG activity in
differentiate an image artifact from a true tumor lesions with normal surrounding tissues
lesion. and organs. This is a purely qualitative
Focal FDG uptake does not necessarily approach, which is unavoidably subjective and
translate to neoplastic involvement, particu- does not allow for detection of subtle changes
larly if the level of uptake is mild to moderate, in tumor uptake. A more precise method is
since nonmalignant conditions such as infec- measurement of a tumor-to-background (i.e.,
tious and inflammatory conditions can demon- normal tissue) ratio (TBR). This is a semiquan-
strate FDG avidity (63-65). These conditions titative method, and has some limitations. For
include pyogenic abscesses, tuberculosis, coc- example, even with a constant 18F-FDG uptake
cidioidomycosis, aspergillosis, histoplasmosis, within the tumor, TBR values could falsely
sarcoidosis, postradiation inflammatory changes, change due to variations in the surrounding
and postoperative wound healing (4). In this normal tissue activity caused by changes in
context, inflammatory changes secondary to glucose level, time of scanning, or therapeutic
local tumor ablation procedures such as cryoa- effect.
blation, radiofrequency ablation, and ethanol Today, the most widely accepted semiquanti-
injection may also lead to false-positive FDG tative method for assessment of treatment
uptake (discussed later in this chapter). response by PET is the standardized uptake
The strength of PET scanning relies on its value (SUY), which is defined as the ratio of
high negative predictive value. If there is no FDG concentration in the tumor to the mean
FDG uptake within a lesion that is at least 7 mm value in the whole body (injected dose divided
or greater in size, it is most likely not malignant. by patient weight in kilograms) (82-84). This
Exceptions to this rule include carcinoid tumor, value has an advantage over TBR in that no
bronchioloalveolar carcinoma, some renal cell measurement of normal tissue is required.
cancers, prostate cancer, hepatocellular carci- However, SUY also may be significantly af-
noma, and cancers with myxoid features. These fected by changes in glucose level, time delay
cancers may exhibit low or no FDG uptake between injection and scanning, or therapy
(4,66-78). effect. Changes in SUY of less than 25%
between scans are not normally considered to
be significant (81,85).
PET Imaging: Methodology
In the longitudinal evaluation of patients with
FDG-PET, attention should be paid to strict Interventional Radiology and
adherence to protocol details such as adequate PET: Clinical Considerations
fasting, glycemic status, time of imaging,
acquisition and quantitation techniques, and Assessment of the therapeutic outcomes of
minimization of subcutaneous dose infiltra- interventional radiology procedures is classi-
tion. These are important factors to consider cally based on bidimensional changes in a lesion
because differences in glycemic or insulin levels size, density, and enhancement changes on CT,
may significantly alter the biodistribution of or signal enhancement characteristics on MRI
FDG. Dose extravasation can result in sig- before and after the treatment. However, such
nificant image artifacts, which may impair anatomic assessments have limitations in cases
quantification accuracy and may result in non- in which residual masses often are seen during
pathologic focal uptake in the lymphatic and after treatment, for example in lymphomas.
system, which may be difficult to distinguish In these cases, differentiation between residual
from malignancy. viable tumor tissue and scar tissue is often
124 A.D. Van den Abbeele et al

limited based on anatomic imaging (e.g., CT, larly helpful to anatomically localize pathologic
MRI) modalities alone. In these circumstances, FDG uptake in retroperitoneal masses and
functional· imaging demonstrates useful com- abdominal or pelvic wall lesions (88). Registra-
plementarity to anatomic imaging and can help tion errors, however, occur because of diffi-
in determining appropriate levels of cancer culties in proper patient repositioning when
treatment in patients. Since FDG-PET scanning imaging with the second modality.
can reliably detect the presence of viable tumor In an attempt to avoid these errors, inte-
within residual masses that are 7 mm or greater grated systems capable of sequentially acquir-
in size, it can help to differentiate residuall ing both PET and CT data in a single imaging
recurrent tumor from posttherapeutic inflam- session have been designed (89-91). The first
matory changes or scar. such system was developed at the University of
The basic prerequisite for use of FDG-PET Pittsburgh and consisted of a spiral CT scanner
in the evaluation of therapeutic response is the integrated with a rotating partial-ring PET
demonstration of FDG avidity within the lesion scanner. Today, combined PETtCT scanners are
of interest (i.e., malignancy) prior to treatment. available from multiple vendors and will likely
Therefore, the usual protocol for use of FDG- dominate the market for PET systems in the
PET in therapeutic evaluation should include a near future. Image fusion obtained on the
baseline scan obtained before the interven- current generation of integrated scanners is less
tional procedure and follow-up scan(s) per- prone to error due to body positioning, but
formed after completion of the therapeutic some inaccuracies still exist due to, for example,
procedure. The timing of the follow-up scan differences in respiratory chest movement
depends on several factors, including the nature between fast CT and slower PET acquisi-
of the treated lesion, the type of the applied tions, or patient motion and bowel activity
therapeutic modality, and the findings of other distribution over the course of the two scans
imaging modalities. The use of FDG-PET in the (92-95).
assessment of tumor ablation in various cancers Nevertheless, early experiences gathered on
is discussed later in this chapter. a wide range of malignancies including lung,
colorectal, head and neck, and ovarian cancer,
as well as lymphoma and melanoma, have been
very positive (96-98). Integrated diagnostic
Combined Multimodality systems will add value in providing not only a
Imaging more accurate diagnosis and localization of the
hypermetabolic focus seen on PET (88,99,100),
As much as FDG-PET can be helpful in the but also in allowing PETtCT-guided interven-
assessment of the viability of a lesion of inter- tions such as aspiration biopsy for lesion
est, the absence of detailed anatomic infor- characterization or transcatheter tumor
mation can lead to diagnostic dilemmas in the chemoembolization, radiofrequency, and other
evaluation of complex anatomic regions. In ablation techniques.
practice, interpretation is performed by visually
correlating the PET scans with high-quality
anatomic images provided by CT or magnetic Preliminary Clinical Experience
resonance (MR) scans (86).
Functional-morphologic correlation may be with FDG-PET in Tumor
assisted by digital image fusion of indepen- Ablation in Our Practice
dently performed PET and CT images. Results
of several studies have shown the superior clin- Over the past several years, FDG-PET imaging
ical value of digitally fused images in patients has become widely used in the diagnosis and
with brain, lung, and abdominal tumors and for staging of a variety of malignant tumors and as
radiation therapy planning (87,88). Such digi- a tool for follow-up of these tumors to assess
tally fused images are reported to be particu- the response to treatment and disease progres-
10. Positron Emission Tomography Imaging for Tumor Ablation 125

sion. This section discusses our clinical ex- should be noted that the standard clinical onco-
perience with the use of FDG-PET imaging logic PET protocol in our Department calls for
for follow-up in patients undergoing tumor imaging the whole body from the base of the
ablation. skull to the proximal thighs. The preprocedure
At our institution, patients referred for MRI tends to scan a more restricted region
tumor ablation are imaged before and after (e.g., liver only, pelvis only), and for that reason
ablation by standard anatomic methods, usually can be expected to be less effective for staging.
by MRI, often supplemented by CT. Occasion- Preprocedure PET can provide valuable
ally, only CT imaging is used if MRI is not fea- staging information. The discovery of previ-
sible. As FDG-PET imaging has become more ously undetected metastatic sites in a patient
commonly used, many of thes€ patients have with an apparent solitary focus may modify the
had pre- and/or postprocedure PET scans. Of plans for ablation, or at least alter the nature of
approximately 160 ablation procedures done the preprocedure counseling of the patient. The
since we began performing PET scans, 21 postablation follow-up studies are of clinical
patients undergoing these procedures have had interest for assessing the effectiveness of the
FDG-PET scans both before and after their procedure in destroying the tumor, for evaluat-
ablation procedures. All 21 patients had stan- ing the presence of residual or recurrent
dard anatomic imaging before and after their disease, and for assessing interval change in
procedures, so we have a basis for qualitative other known sites, or development of new
comparison of the information provided by disease sites (Fig. 10.1) (101,102).
anatomic and functional imaging in ablation In five of these nine patients, both follow-up
cases. The following discussion is based on clin- PET and MRI demonstrated recurrent disease
ical interpretations, in which the imaging scan at the ablation site. In another three of these
readers had access to any and all clinical infor- nine patients, follow-up PET and MRI showed
mation, including the results of other imaging no evidence of recurrence at the ablation site,
techniques, and is intended to illustrate the clin- although in one the follow-up PET showed
ical possibilities and some of the potential pit- extensive new metastatic disease elsewhere in
falls of FDG-PET imaging in the context of liver, nodes, skeleton, and lungs. Finally, in one
tumor ablation. of these nine colorectal cancer patients, a post-
procedure PET 3 months posttherapy detected
mildly increased uptake at the site of one of
Colorectal Cancer the ablated lesions, which was indeterminate
The largest subgroup of these patients, nine of for either inflammatory or residual disease.
the 21, had colorectal cancer. Six of these However, subsequent PET scans showed
patients had metastatic disease to the liver that further resolution of tracer uptake at this site
was the target for ablation. One patient had in spite of documenting definite progression of
adrenal and lung metastases in addition to liver disease elsewhere. Thus, FDG tracer uptake is
metastases. The remaining two patients had not specific for tumor, and intense tracer
pelvic masses, which were the target lesions. uptake can be seen at sites of inflammation,
One patient study in our group of colorectal which may occur at postablation sites.
cancer patients was limited by a hyperglycemic
state.
Typically in this disease setting, the prepro-
Lung Cancer
cedure MRI and PET scans were in agreement Six of the 21 patients who underwent ablation
regarding the disease in the target lesion(s).All with both pre- and postprocedure PET imaging
of the intended target lesions were detectable had lung cancer of various types. Three of these
by both modalities. Other metastatic lesions, had metastatic lesions to the liver that were the
some definite and some questionable, were target lesions for ablation, while the others had
detected on the preprocedure studies in four of lung or chest-wall lesions that were not
these patients by PET and in two by MRI. It amenable to surgery. In all of these cases, the
126 A.D. Van den Abbeele et al

A B
FIGURE 10.1. Colon cancer metastatic to liver, lung, (B) Postprocedure study. The left lung and adrenal
and left adrenal. Left anterior oblique (LAO) lesions show posttherapeutic inflammatory changes,
projection images (MIP). (A) Preprocedure study. although residual tumor cannot be excluded. The
Metastases are seen in the right lobe of the liver, left metastatic disease to the liver has progressed. A new
lung base, and left adrenal. Note physiologic tracer small focus that projects just below the heart is a new
excretion in kidneys, right ureter, and bladder. metastasis located in the right lung base.

target lesion was detected preprocedure by concordant, suggesting recurrent or residual


both anatomic and functional imaging. tumor.
In one patient with squamous cell lung There were three patients with lung pri-
cancer metastatic to liver, initial and follow-up maries in whom the target lesion for ablation
PET scans identified a hilar nodal metastasis was in the chest. One of these patients had a
that was not seen on the liver MRI. In another lung tumor seen on both initial MR and PET.
patient with non-small-cell lung cancer with The repeat PET 1 month after the procedure
several metastases to liver, the postprocedure showed residual disease at the ablation site and
PET and MRI both showed residual lesions. also a new metastasis in the pelvis, which was
The PET result was interpreted as showing not, of course, seen on the postprocedure chest
posttherapeutic inflammation at a treated site, CT. Another patient had non-small-cell lung
while the MRI scan was interpreted as showing cancer with a large right apical mass with chest
residual disease. However, a subsequent repeat wall invasion, and a right hilar mass, both of
PET scan confirmed disease progression at this which were evident on the preprocedure PET
site. In a third patient with lung cancer, a single and chest MRI, and were consistent with neo-
5-cm liver metastasis was identified and treated. plasm by PET. This patient was not a candidate
The PET obtained 3 months after the proce- for surgery, and radiofrequency ablation of
dure showed only slight interval decrease in the lung mass was undertaken with palliative
tracer uptake in the lesion, and was consistent intent. A postprocedure PET at 6 weeks
with residual tumor. The MRI appearance was demonstrated a central photopenic region at
10. Positron Emission Tomography Imaging for Tumor Ablation 127

the site of the ablation, with a rim of surround- nodule anterior to the main lesion, suggesting
ing increased FDG uptake. The latter appear- recurrent disease.
ance could have been caused either by residual
tumor at the boundaries of the treated region,
or alternately by postprocedure inflammatory
Other Tumors
change, although the former explanation was Ovarian Cancer
favored. A subsequent follow-up CT obtained
One patient had ovarian cancer metastatic to
5 weeks after the PET did demonstrate inter-
the abdomen, pelvis, and liver surface that was
val increase in the size of the mass, confirming
detected by PET, although two small liver
progression of the tumor.
lesions seen on CT (less than 1 cm) were not
The differential diagnosis of residual tumor
visualized on PET, probably because of the
versus inflammatory changes at a site of
small size of the lesions.
increased or residual FDG uptake may be chal-
In this patient, the preprocedure MRI showed
lenging in the early postprocedure phase since
at least four metastatic lesions to the liver.
posttherapeutic inflammatory changes also can
The patient underwent radiofrequency/ethanol
cause increased uptake of tracer particularly
ablation of two of these lesions for symptomatic
soon after therapy. This phenomenon has been
relief. The postprocedure MRI showed treat-
described during radiation therapy (4,45,103-
ment effect in some of the lesions, and growth of
106). Since qualitative readings of PET scans can
nontreated lesions. A postprocedure PET per-
be reader-dependent, training and experience in
formed 5 months later showed interval complete
PET imaging interpretation help nuclear medi-
resolution of one treated lesion, residual disease
cine physicians and radiologists become familiar
at another, and interval development of a new
with the various patterns of inflammatory versus
lesion in Morison's pouch.
tumoral uptake. Dual time point FDG-PET
imaging at 45 to 70 minutes and at approximately
Cholangiocarcinoma
90 minutes postinjection of the tracer also has
been proposed to help in the differentiation In a patient with cholangiocarcinoma, a pre-
of benign from malignant disease (107,108). procedure MRI showed a 16-cm central hepatic
Continuous increase in SUV was seen in mass with vascular encasement. The initial PET
malignant lesions, while SUVs of benign lesions showed a large central liver lesion with central
or inflammation decreased over time, or photopenia, and possible bone lesions were
remained stable. Follow-up scans 4 to 6 weeks or detected.
later after the procedure also can be helpful Larger tumors often have a shell-like ap-
since inflammatory changes may continue to pearance on PET, with relative photopenia
resolve with time after the procedure, assuming centrally. Reasons for this observation could
that the postprocedure course is not complicated include central necrosis of the tumor, with a
by infection or other unusual circumstances. lack of viable cells in the photopenic regions, or
However, long-term residual uptake has been relatively poor blood supply and tracer delivery
observed, particularly after proton therapy to these regions. This fact can somewhat com-
(106). plicate the interpretation of a tumor ablation
The last patient in our study population had case, in which the therapy is applied to the
lung cancer, with a large left lung lesion visible center of the lesion, which in a big lesion may
on CT and PET. Follow-up PET, about 1 month have shown little tracer uptake to begin with.
after the ablation, showed a photopenic defect In the case of this patient, the follow-up MRI
with mild increased uptake at the rim thought at 1 week postprocedure showed no change in
to be inflammatory and two distinct small the appearance of the tumor; PET at 5 weeks
intense foci, which could be residual disease showed a larger region of central photopenia in
(Fig. 10.2). Follow-up CT performed 2 months the tumor, but peripheral tracer uptake. This
later showed treatment changes in the main was interpreted as showing partial response to
lesion, but an interval increase in a separate the ablation therapy.
128 A.D. Van den Abbeele et al

A B
FIGURE 10.2. (A) Preprocedure study. A single lesion shows central photopenia in the lesion, with
coronal image showing an intensely FDG-avid surrounding mild uptake consistent with postproce-
non-small-celllung tumor in the left lung. Note the dure inflammatory changes and a slightly more
right hip prosthesis. (B) Postprocedure study at 1 intense focus at the upper margin, suspicious for
month. A single coronal image through the treated residual disease.

Gastric Cancer procedure, the patient had been started on new


chemotherapy, which most likely was respon-
One patient had gastric cancer metastatic to the
sible for the improvement in all lesions.
gastrohepatic ligament and the retroperi-
toneum around both the celiac axis and superior
Gastrointestinal Stromal Tumor (GIST)
mesenteric artery. These lesions were visible on
both MRI and PET prior to the procedure. A One patient with GIST had multiple metastatic
PET obtained 2 months after the procedure liver and abdominal lesions that were pro-
demonstrated central photopenia in the ablated gressing on therapy. A preprocedure PET
lesion, but otherwise interval progression of showed recurrence of GIST in previously
disease. An MRI obtained after another 3 treated locations in the right lobe of the liver,
months showed overall improvement. However, with a possible new lesion in the left lobe
according to the report of that study, some non- despite ongoing therapy with imatinib mesylate
ablated sites improved as well. Following the (Gleevec).A postprocedure PET done 6 weeks
10. Positron Emission Tomography Imaging for Tumor Ablation 129

later showed interval resolution of the in- formed at a lower cost and with less associated
creased uptake in the treated lesions, and no morbidity than other interventions. Radiologic
definite evidence of metabolically active tumor. assessment of bidimensional changes in tumor
A CT performed at 6 weeks provided confir- size is still largely considered a "gold standard"
matory evidence that the treated lesions for the evaluation of therapeutic response in
showed no recurrence. such patients. However, there is rapidly increas-
ing evidence that functional imaging with
Tumors with Variable FDG Avidity FDG-PET has a significant advantage in the
evaluation of response to therapy, particularly
Renal Cell Carcinoma in the evaluation of residual masses.
Not all tumor types show increased avidity for
FDG, so FDG-PET cannot be used to follow Acknowledgment
such cases if there is no FDG avidity within the
tumor site in the baseline scan. For example, The authors gratefully acknowledge the valu-
one patient with a history of metastatic ovarian able assistance of Clay H. Holdsworth, Ph.D.,
cancer had a large left renal mass, presumably and Richard 1. Tetrault, CNMT, for their assis-
metastatic, for which ablation was being con- tance in reviewing this chapter.
sidered. The preprocedure PET demonstrated
the multiple FDG-avid metastatic foci of References
ovarian cancer in the abdomen, but no focal
increased uptake in the large renal lesion. A 1. Maisey MN, Wahl RL, Barrington SF. Atlas
subsequent biopsy showed the renal mass to be of Clinical Positron Emission Tomography.
a renal cell carcinoma. London: Arnold, 1999:346.
Some primary renal cell carcinomas can be 2. Brown RS, Wahl RL. Overexpression of Glut-l
glucose transporter in human breast cancer.
negative on FDG-PET (69), but others are not,
An immunohistochemical study. Cancer 1993;
and a baseline FDG-PET is helpful to deter- 72(10):2979-2985.
mine if PET could be useful or not. In this par- 3. Brown RS, et al. Expression of hexokinase II
ticular case, FDG-PET was a valuable tool to and Glut-l in untreated human breast cancer.
assess the patient's ovarian cancer, but not for Nucl Med Bioi 2002;29(4):443-453.
the second renal cell primary. 4. Wieler H, Coleman R. PET in Clinical Oncol-
ogy. Darmstadt: Steinkopff Verlag, 2000:422.
Hepatocellular Carcinoma 5. Gambhir SS, Shepherd JE, Shah BD, et al. Ana-
lytical decision model for the cost-effective
Hepatocellular carcinoma (HCC) is a tumor management of solitary pulmonary nodules.
with variable avidity for FDG (74-77). One J Clin OncoI1998;16(6):2113-2125.
patient had a known 2.7-cm HCC in the infe- 6. Gambhir SS, Czernin J, Schwimmer J,
rior posterior segment of the right lobe. This Silverman DH, Coleman RE, Phelps ME.
lesion was seen on the MRI, but was not A tabulated summary of the FDG PET litera-
detected on the PET done the next day. Again, ture. J Nucl Med 2001;42(5 suppl):IS-93S.
baseline PET may help determine if this modal- 7. Huebner RH, Park KC, Shepherd JE, et al. A
ity might be useful in the follow-up of these meta-analysis of the literature for whole-body
FDG PET detection of recurrent colorectal
patients.
cancer. J Nucl Med 2000;41(7):1177-1189.
8. Kubik-Huch RA, Dorffler W, von Schulthess
GK, et al. Value of (18F)-FDG positron emis-
Conclusion sion tomography, computed tomography, and
magnetic resonance imaging in diagnosing
Interventional radiology techniques typically primary and recurrent ovarian carcinoma. Eur
represent the least invasive definitive diagnos- RadioI2000;10(5):761-767.
tic or therapeutic options available for patients 9. Meta J, Seltzer M, Schiepers C, et al. Impact of
with cancer. These techniques are usually per- 18F-FDG PET on managing patients with col-
130 A.D. Van den Abbeele et al

orectal cancer: the referring physician's per- in the evaluation of patients with residual and
spective. J Nucl Med 2001;42(4):586-590. recurrent Hodgkin's lymphoma. Oncol Rep
10. Park KC, Schwimmer J, Shepherd JE, et al. 2001;8(6):1393-1399.
Decision analysis for the cost-effective man- 21. Mikhaeel NG, Timothy AR, O'Doherty MJ,
agement of recurrent colorectal cancer. Ann Hain S, Maisey MN. 18-FDG-PET as a prog-
Surg 2001;233(3):310-319. nostic indicator in the treatment of aggres-
11. Park KC, Schwimmer J, Gambhir SS. Decision sive non-Hodgkin's lymphoma-comparison
analysis for the cost-effective management of with CT. Leuk Lymphoma 2000;39(5-6):543-
recurrent colorectal cancer. Ann Surg 2002; 553.
235(2):309-310; author reply 10. 22. Pieterman RM, van Putten JW, Meuzelaar n, et
12. Schwimmer J, Essner R, Patel A, et al. A review al. Preoperative staging of non-small-cell lung
of the literature for whole-body FDG PET in cancer with positron-emission tomography. N
the management of patients with melanoma. Q Engl J Med 2000;343(4):254-261.
J Nucl Med 2000;44(2):153-167. 23. Berman CG, Clark RA. Positron emission
13. Scott WJ, Shepherd J, Gambhir SS. Cost- tomography in initial staging and diagnosis
effectiveness of FDG-PET for staging non- of persistent or recurrent disease. Curr Opin
small cell lung cancer: a decision analysis. Ann OncoI2000;12(2):132-137.
Thorac Surg 1998;66(6):1876-1883; discussion 24. Lowe VJ, Fletcher JW, Gobar L, et al. Prospec-
83-85. tive investigation of positron emission tomog-
14. Seltzer MA, Yap CS, Silverman DH, et al. The raphy in lung nodules. J Clin OncoI1998;16(3):
impact of PET on the management of lung 1075-1084.
cancer: the referring physician's perspective. 25. Lowe VJ, Naunheim KS. Positron emission
J Nucl Med 2002;43(6):752-756. tomography in lung cancer. Ann Thorac Surg
15. Wu D, Gambhir SS. Positron emission tomog- 1998;65(6):1821-1829.
raphy in diagnosis and management of invasive 26. Lowe VJ, Naunheim KS. Current role of
breast cancer: current status and future per- positron emission tomography in thoracic
spectives. Clin Breast Cancer 2003;4(suppll): oncology. Thorax 1998;53(8):703-712.
S55-S63. 27. Delbeke D. Oncological applications of FDG
16. Yap CS, Seltzer MA, Schiepers C, et al. Impact PET imaging: brain tumors, colorectal cancer,
of whole-body 18F-FDG PET on staging lymphoma and melanoma. J Nucl Med 1999;
and managing patients with breast cancer: the 40(4):591-603.
referring physician's perspective. J Nucl Med 28. Delbeke D. Oncological applications of FDG
2001;42(9):1334-1337. PET imaging. J Nucl Med 1999;40(10):1706-
17. Services DOHH. Coverage and related claims 1715.
processing requirements for positron emission 29. Erasmus n, McAdams HP, Connolly JE.
tomography (PET) scans. Program Memoran- Solitary pulmonary nodules: part II. Evaluation
dum Intermediaries/Carriers. CMS-Pub. 60AB of the indeterminate nodule. Radiographics
(AB-02-065).2002:1-4. 2000;20(1):59-66.
18. Kinkel K, Lu Y, Both M, Warren RS, Thoeni RF. 30. Erasmus n, Connolly JE, McAdams HP, Roggli
Detection of hepatic metastases from cancers VL. Solitary pulmonary nodules: part I. Mor-
of the gastrointestinal tract by using noninva- phologic evaluation for differentiation of
sive imaging methods (US, CT, MR imaging, benign and malignant lesions. Radiographics
PET): a meta-analysis. Radiology 2002;224(3): 2000;20(1):43-58.
748-756. 31. Coleman RE. PET in lung cancer. J Nucl Med
19. Jerusalem G, Beguin Y, Fassotte MF, et al. 1999;40(5):814-820.
Whole-body positron emission tomography 32. Erasmus n, McAdams Hp, Patz EF, Jr. Non-
using 18F-fluorodeoxyglucose for posttreat- small cell lung cancer: FDG-PET imaging.
ment evaluation in Hodgkin's disease and non- J Thorac Imaging 1999;14(4):247-256.
Hodgkin's lymphoma has higher diagnostic and 33. Rinne D, Baum RP, Hor G, Kaufmann R.
prognostic value than classical computed Primary staging and follow-up of high risk
tomography scan imaging. Blood 1999;94(2): melanoma patients with whole-body 18F-
429-433. fluorodeoxyglucose positron emission tomog-
20. Dittmann H, Sokler M, Kollmannsberger C, et raphy: results of a prospective study of 100
al. Comparison of 18FDG-PET with CT scans patients. Cancer 1998;82(9):1664-1671.
10. Positron Emission Tomography Imaging for Tumor Ablation 131

34. Lowe VJ, Boyd JH, Dunphy FR, et al. Surveil- metastases: correlation with tumor response
lance for recurrent head and neck cancer using to fluorouracil. J Clin Oncol 1996;14(3):700-
positron emission tomography. J Clin Oncol 708.
2000;18(3):651-658. 45. Haberkorn U, Strauss LG, Dimitrakopoulou A,
35. Skehan SJ, Brown AL, Thompson M, Young JE, et al. PET studies of fluorodeoxyglucose
Coates G, Nahmias e. Imaging features of metabolism in patients with recurrent colorec-
primary and recurrent esophageal cancer at tal tumors receiving radiotherapy. J Nucl Med
FDG PET. Radiographies 2000;20(3):713- 1991;32(8):1485-1490.
723. . 46. Ichiya Y, Kuwabara Y, Otsuka M, et al. Assess-
36. Luketich JD, Friedman DM, Weigel TL, et al. ment of response to cancer therapy using
Evaluation of distant metastases in esophageal fluorine-18-fluorodeoxyglucose and positron
cancer: 100 consecutive positron emission emission tomography. J Nucl Med 1991;32(9):
tomography scans. Ann Thorac Surg 1999;68(4): 1655-1660.
1133-1136; discussion 1136-1137. 47. Okazumi S, Isono K, Enomoto K, et al. Evalu-
37. Zimmer LA, McCook B, Meltzer C, et al. Com- ation of liver tumors using fluorine-18-fluo-
bined positron emission tomography/computed rodeoxyglucose PET: characterization of tumor
tomography imaging of recurrent thyroid and assessment of effect of treatment. J Nucl
cancer. Otolaryngol Head Neck Surg 2003; Med 1992;33(3):333-339.
128(2):178-184. 48. Reisser C, Haberkorn U, Dimitrakopoulou-
38. Frilling A Tecklenborg K Gorges R, Weber Strauss A, Seifert E, Strauss LG. Chemo-
F, Clausen M, Broelsch Ee. Preoperative therapeutic management of head and neck
diagnostic value of [(18)F] fluorodeoxyglucose malignancies with positron emission tomo-
positron emission tomography in patients graphy. Arch Otolaryngol Head Neck Surg
with radioiodine-negative recurrent well- 1995;121(3):272-276.
differentiated thyroid carcinoma. Ann Surg 49. Wahl RL, Zasadny K, Helvie M, Hutchins GD,
2001;234(6): 804-811. Weber B, Cody R. Metabolic monitoring of
39. Helal BO, Merlet P, Toubert ME, et al. Clinical breast cancer chemohormonotherapy using
impact of (18)F-FDG PET in thyroid carci- positron emission tomography: initial evalua-
noma patients with elevated thyroglobulin tion. J Clin OncoI1993;11(11):2101-2111.
levels and negative (131)1 scanning results after 50. Schulte M, Brecht-Krauss D, Werner M, et al.
therapy. J Nucl Med 2001;42(10):1464-1469. Evaluation of neoadjuvant therapy response of
40. Yeo JS, Chung JK, SO Y, et al. F-18- osteogenic sarcoma using FDG PET. J Nucl
fluorodeoxyglucose positron emission tomo- Med 1999;40(10):1637-1643.
graphy as a presurgical evaluation modality for 51. Minn H, Lapela M, Klemi PJ, et al. Prediction
1-131 scan-negative thyroid carcinoma patients of survival with fluorine-18-fluoro-deoxyglu-
with local recurrence in cervical lymph nodes. cose and PET in head and neck cancer. J Nucl
Head Neck 2001;23(2):94-103. Med 1997;38(12):1907-1911.
41. Hustinx R, Benard F, Alavi A. Whole-body 52. Alial AS, Dulguerov P, Allaoua M, et al. Stan-
FDG-PET imaging in the management of dardized uptake value of 2-[(18)F] fluoro-2-
patients with cancer. Semin Nucl Med 2002; deoxy-D-glucose in predicting outcome in head
32(1):35-46. and neck carcinomas treated by radiotherapy
42. Bassa P, Kim EE, Inoue T, et al. Evaluation of with or without chemotherapy. J Clin Oncol
preoperative chemotherapy using PET with 2002;20(5):1398-1404.
fluorine-18-fluorodeoxyglucose in breast can- 53. Eary JF, Krohn KA. Positron emission tomo-
cer. J Nucl Med 1996;37(6):931-938. graphy: imaging tumor response. Eur J Nucl
43. Berlangieri SU, Brizel DM, Scher RL, et al. Med 2000;27(12):1737-1739.
Pilot study of positron emission tomography in 54. Folpe AL, Lyles RH, Sprouse JT, Conrad EU,
patients with advanced head and neck cancer III, Eary JF. (F-18) fluorodeoxyglucose positron
receiving radiotherapy and chemotherapy. emission tomography as a predictor of patho-
Head Neck 1994;16(4):340-346. logic grade and other prognostic variables in
44. Findlay M, Young H, Cunningham D, et al. bone and soft tissue sarcoma. Clin Cancer Res
Noninvasive monitoring of tumor metabolism 2000;6(4):1279-1287.
using fluorodeoxyglucose and positron emis- 55. Wong RJ, Lin DT, Schoder H, et al. Diagnostic
sion tomography in colorectal cancer liver and prognostic value of [(18)F]fluorodeoxyglu-
132 A.D. Van den Abbeele et al

cose positron emission tomography for recur- 65. Yoon SN, Park CH, Kim MK, Hwang KH, Kim
rent head and neck squamous cell carcinoma. S. False-positive F-18 FDG gamma camera
J Clin Oncol 2002;20(20):4199-4208. positron emission tomographic imaging result-
56. Spaepen K, Mortelmans L. Evaluation of treat- ing from inflammation of an anterior mediasti-
ment response in patients with lymphoma using nal mass in a patient with non-Hodgkin's
[18F]FDG-PET: differences between non- lymphoma. Clin Nucl Med 2001;26(5):461-
Hodgkin's lymphoma and Hodgkin's disease. Q 462.
J Nucl Med 2001;45(3):269-273. 66. Erasmus n, McAdams Hp, Patz EF, Jr.,
57. Spaepen K, Stroobants S, Dupont P, et al. Can Coleman RE, Ahuja V, Goodman Pc. Evalua-
positron emission tomography with [(18)F]- tion of primary pulmonary carcinoid tumors
fluorodeoxyglucose after first-line treatment using FDG PET. AJR Roentgenol 1998;170(5):
distinguish Hodgkin's disease patients who 1369-1373.
need additional therapy from others in 67. Rege SD, Hoh CK, Glaspy JA, et al. Imaging
whom additional therapy would mean avoid- of pulmonary mass lesions with whole-body
able toxicity? Br J Haematol 2001;115(2): positron emission tomography and fluorode-
272-278. oxyglucose. Cancer 1993;72(1):82-90.
58. Spaepen K, Stroobants S, Dupont P, et al. Prog- 68. Hoh CK, Seltzer MA, Franklin J, deKernion JB,
nostic value of positron emission tomography Phelps ME, Belldegrun A. Positron emission
(PET) with fluorine-18 fluorodeoxyglucose tomography in urological oncology. J Urol
([18F]FDG) after first-line chemotherapy in 1998;159(2):347-356.
non-Hodgkin's lymphoma: is [18F]FDG-PET a 69. Miyakita H, Tokunaga M, Onda H, et al. Sig-
valid alternative to conventional diagnostic nificance of 18F-fluorodeoxyglucose positron
methods? J Clin Oncol 2001;19(2):414-419. emission tomography (FDG-PET) for detec-
59. Jerusalem G, Warland V, Najjar F, et al. Whole- tion of renal cell carcinoma and immunohisto-
body 18F-FDG PET for the evaluation of chemical glucose transporter 1 (GLUT-I)
patients with Hodgkin's disease and non- expression in cancer. Int J Urol2oo2;9(1):15-18.
Hodgkin's lymphoma. Nucl Med Commun 70. Hofer C, Kubler H, Hartung R, Breul J, Avril N.
1999;20(1):13-20. Diagnosis and monitoring of urological tumors
60. Naumann R, Vaic A, Beuthien-Baumann B, et using positron emission tomography. Eur Urol
al. Prognostic value of positron emission 2001;40(5):481-487.
tomography in the evaluation of post-treatment 71. Effert PJ, Bares R, Handt S, Wolff JM, Bull U,
residual mass in patients with Hodgkin's Jakse G. Metabolic imaging of untreated
disease and non-Hodgkin's lymphoma. Br J prostate cancer by positron emission tomo-
HaematoI2001;1l5(4):793-800. graphy with 18-fluorine-Iabeled deoxyglucose.
61. de Wit M, Bohuslavizki KH, Buchert R, J Urol 1996;155(3):994-998.
Bumann D, Clausen M, Hossfeld DK. 18FDG- 72. Hofer C, Laubenbacher C, Block T, Breul J,
PET following treatment as valid predictor for Hartung R, Schwaiger M. Fluorine-18-fluo-
disease-free survival in Hodgkin's lymphoma. rodeoxyglucose positron emission tomography
Ann Oncol 2001;12(1):29-37. is useless for the detection of local recurrence
62. Becherer A, Mitterbauer M, Jaeger U, et al. after radical prostatectomy. Eur Urol 1999;
Positron emission tomography with [18F]2- 36(1):31-35.
fluoro-D-2-deoxyglucose (FDG-PET) predicts 73. Seltzer MA, Barbaric Z, Belldegrun A, et al.
relapse of malignant lymphoma after high- Comparison of helical computerized tomogra-
dose therapy with stem cell transplantation. phy, positron emission tomography and mono-
Leukemia 2002;16(2):260-267. clonal antibody scans for evaluation of lymph
63. Bakheet SM, Saleem M, Powe J, AI-Amro A, node metastases in patients with prostate spe-
Larsson SG, Mahassin Z. F-18 fluorodeoxyglu- cific antigen relapse after treatment for local-
cose chest uptake in lung inflammation and ized prostate cancer. J Urol 1999;162(4):1322-
infection. Clin Nucl Med 2000;25(4):273- 1328.
278. 74. Dimitrakopoulou-Strauss A, Gutzler F, Strauss
M. Wolf G, Aigner RM, Schwarz T. Pathologic LG, et al. [PET studies with C-ll ethanol in
uptake in F-18 FDG positron emission tomo- intratumoral therapy of hepatocellular carcino-
graphy of the residuals of a surgically removed mas]. Radiologe 1996;36(9):744-749.
needle abscess. Clin Nucl Med 2002;27(6):439- 75. Wudel LJ, Jr., Delbeke D, Morris D, et al. The
440. role of [18F]fluoTodeoxyglucose positron emis-
10. Positron Emission Tomography Imaging for Tumor Ablation 133

sion tomography imaging in the evaluation of 85. Hallett WA, Marsden PK, Cronin BF, O'Do-
hepatocellular carcinoma. Am Surg 2003;69(2): herty MJ. Effect of corrections for blood
117-124, discussion 24-26. glucose and body size on [18F]FDG PET stan-
76. Iwata Y, Shiomi S, Sasaki N, et al. Clinical use- dardised uptake values in lung cancer. Eur J
fulness of positron emission tomography with Nucl Med 2001;28(7):919-922.
fluorine-18-fluorodeoxyglucose in the diagnosis 86. Zimny M, Wildberger JE, Cremerius D, et al.
of liver tumors. Ann Nucl Med 2000;14(2):121- [Combined image interpretation of computed
126. tomography and hybrid PET in head and
77. Schroder 0, Trojan J, Zeuzem S, Baum RP. neck cancer.] Nuklearmedizin 2002;41(1):14-
[Limited value of fluorine-18-fluorodeoxyglu- 21.
cose PET for the differential diagnosis of focal 87. Caldwell CB, Mah K, Dng YC, et al. Observer
liver lesions in patients with chronic hepatitis C variation in contouring gross tumor volume in
virus infection]. Nuklearmedizin 1998;37(8): patients with poorly defined non-small-celllung
279-285. tumors on CT: the impact of 18FDG-hybrid
78. Schwarzbach MH, Dimitrakopoulou-Strauss A, PET fusion. Int J Radiat Oncol BioI Phys
Mechtersheimer G, et al. Assessment of soft 2001;51(4):923-931.
tissue lesions suspicious for liposarcoma by 88. Schaffier GJ, Groell R, Schoellnast H, et al.
F18-deoxyglucose (FDG) positron emission Digital image fusion of CT and PET data sets-
tomography (PET). Anticancer Res 2001;21(5): clinical value in abdominaUpelvic malignancies.
3609-3614. J Comput Assist Tomogr 2000;24(4):644-
79. Hoekstra CJ, Paglianiti I, Hoekstra OS, et al. 647.
Monitoring response to therapy in cancer using 89. Beyer T, Townsend DW, Brun T, et al. A com-
[18F]-2-fluoro-2-deoxy-D-glucose and positron bined PET/CT scanner for clinical oncology.
emission tomography: an overview of different J Nucl Med 2000;41(8):1369-1379.
analytical methods. Eur J Nucl Med 2000;27(6): 90. Townsend DW, Cherry SR. Combining ana-
731-743. tomy and function: the path to true image
80. Weber WA, Ziegler SI, Thodtmann R, fusion. Eur RadioI2001;11(1O):1968-1974.
Hanauske AR, Schwaiger M. Reproducibility 91. Cook GJ, Ott RJ. Dual-modality imaging. Eur
of metabolic measurements in malignant RadioI2001;11(10):1857-1858.
tumors using FDG PET. J Nucl Med 1999; 92. Osman MM, Cohade C, Nakamoto Y, Marshall
40(11):1771-1777. LT, Leal JP, Wahl RL. Clinically significant inac-
81. Young H, Baum R, Cremerius D, et al. Mea- curate localization of lesions with PET/CT: fre-
surement of clinical and subclinical tumour quency in 300 patients. J Nucl Med 2003;44(2):
response using [18F]-fluorodeoxyglucose and 240---243.
positron emission tomography: review and 1999 93. Osman MM, Cohade C, Nakamoto Y, Wahl RL.
EORTC recommendations. European Organi- Respiratory motion artifacts on PET emission
zation for Research and Treatment of Cancer images obtained using CT attenuation correc-
(EORTC) PET Study Group. Eur J Cancer tion on PET-CT. Eur J Nucl Med Mol Imaging
1999;35(13):1773-1782. 2003;30(4):603-606.
82. Ahuja Y, Coleman RE, Herndon J, Patz EF, Jr. 94. Beyer T, Antoch G, Blodgett T, Freudenberg LF,
The prognostic significance of fiuorodeoxyglu- Akhurst T, Mueller S. Dual-modality PET/CT
cose positron emission tomography imaging for imaging: the effect of respiratory motion on
patients with nonsmall cell lung carcinoma. combined image quality in clinical oncology.
Cancer 1998;83(5):918-924. Eur J Nucl Med Mol Imaging 2003;30(4):588-
83. Schelling M, Avril N, Nahrig J, et al. Positron 596.
emission tomography using [(18)F]Fluorode- 95. Nakamoto Y, Tatsumi M, Cohade C, Osman M,
oxyglucose for monitoring primary chemother- Marshall LT, Wahl RL. Accuracy of image
apy in breast cancer. J Clin Oncol 2000;18(8): fusion of normal upper abdominal organs visu-
1689-1695. alized with PET/CT. Eur J Nucl Med Mol
84. Saunders CA, Dussek JE, O'Doherty MJ, Imaging 2003;30(4):597-602.
Maisey MN. Evaluation of fluorine-18- 96. Israel 0, Keidar Z, Iosilevsky G, Bettman L,
fiuorodeoxyglucose whole body positron Sachs J, Frenkel A. The fusion of anatomic
emission tomography imaging in the staging of and physiologic imaging in the management
lung cancer. Ann Thorac Surg 1999;67(3):790--- of patients with cancer. Semin Nucl Med
797. 2001;31(3):191-205.
134 A.D. Van den Abbeele et al

97. Martinelli M, Townsend D, Meltzer C, Ville- 103. Hautzel H, Muller-Gartner HW. Early changes
magne VV. 7. Survey of Results of Whole Body in fluorine-18-FDG uptake during radiother-
Imaging Using the PET/CT at the University of apy. J Nucl Med 1997;38(9):1384-1386.
Pittsburgh Medical Center PET Facility. Clin 104. Engenhart R, Kimmig B, Hover KH, Strauss
Positron Imaging 2000;3(4):161. LG, Lorenz WJ, Wannenmacher M. [Photon-
98. D'Amico TA, Wong TZ, Harpole DH, Brown neutron therapy for recurrent colorectal
SD, Coleman RE. Impact of computed cancer-follow up and preliminary results.]
tomography-positron emission tomography Strahlenther OnkoI1990;166(1):95-98.
fusion in staging patients with thoracic malig- 105. Higashi K, Clavo AC, Wahl RL. In vitro
nancies. Ann Thorac Surg 2002;74(1):160-163; assessment of 2-fluoro-2-deoxy-D-glucose, L-
discussion 3. methionine and thymidine as agents to monitor
99. Charron M, Beyer T, Bohnen NN, et at. Image the early response of a human adenocarcinoma
analysis in patients with cancer studied with a cell line to radiotherapy. J Nucl Med 1993;34(5):
combined PET and CT scanner. Clin Nucl Med 773-779.
2000;25 (11):905-910. 106. Fischman AJ, Thornton AF, Frosch MP,
100. Hany TF, Steinert HC, Goerres GW, Buck A, Swearinger B, Gonzalez RG, Alpert NM. FDG
von Schulthess GK. PET diagnostic accuracy: hypermetabolism associated with inflammatory
improvement with in-line PET-CT system: necrotic changes following radiation of menin-
initial results. Radiology 2002;225(2):575-581. gioma. J Nucl Med 1997;38(7):1027-1029.
101. Langenhoff BS. Oyen WJ, Jager GJ, et at. Effi- 107. Zhuang H, Pourdehnad M, Lambright ES, et at.
cacy of fluorine-18-deoxyglucose positron emis- Dual time point 18F-FDG PET imaging for
sion tomography in detecting tumor recurrence differentiating malignant from inflammatory
after local ablative therapy for liver metastases: processes. J Nucl Med 2001;42(9):1412-1417.
a prospective study. J Clin Oncol 2002;20(22): 108. Hustinx R, Smith RJ, Benard F, et at. Dual time
4453--4458. point fluorine-18 fluorodeoxyglucose positron
102. Anderson GS, Brinkmann F, Soulen MC, Alavi emission tomography: a potential method to
A, Zhuang H. FDG positron emission tomog- differentiate malignancy from inflammation
raphy in the surveillance of hepatic tumors and normal tissue in the head and neck. Eur J
treated with radiofrequency ablation. Clin Nucl Nucl Med 1999;26(10):1345-1348.
Med 2003;28(3):192-197.
11
Ultrasound Imaging in Tumor Ablation
Massimo Tonolini and Luigi Solbiati

Diagnostic imaging plays a key role in all steps Detection of Lesions and
of radiofrequency (RF) tumor ablation:
Selection of Patients
1. Detection of lesions and selection of patients
for treatment; A wide range of treatment options currently is
2. Targeting of lesions and guidance of the available for both primary and metastatic liver
procedure; tumors. Therefore, timely d~tection and accu-
3. Immediate assessment of treatment result; rate quantification of neoplastic involvement at
4. Long-term follow-up. the time of diagnosis or during the course of the
disease allows optimal patient management
Conventional, unenhanced ultrasound (US) and treatment selection and ultimately may
is utilized worldwide for screening of liver result in prolonged survival and possible cure.
disease, but variable sensitivity and well-known Adequate local tumor control is feasible
drawbacks limit its role in the staging of by percutaneous RF ablation provided that
liver tumors. Furthermore, sonography repre- correct indications are observed. In most insti-
sents the most commonly used imaging modal- tutions patients with chronic hepatitis or cir-
ity for the guidance of percutaneous ablative rhosis without functional liver decompensa-
treatments owing to its availability, rapidity, tion, portal thrombosis, or extrahepatic tumor
and ease of use. Differentiation of induced spread may undergo RF of one to four or five
necrosis from viable tumor is not possible with dysplastic lesions or hepatocellular carcinoma
baseline and color Doppler sonography, and foci, each not exceeding 4 to 4.5 cm; hepatomas
therefore the immediate and long-term assess- larger than 5 cm usually are treated by means
ment of therapeutic result is usually accom- of combined therapies (transarterial chemoem-
plished by contrast-enhanced helical computed bolization, ethanol injection, RF) (2-4). Effec-
tomography (CT) and magnetic resonance tive ablation of liver metastases requires the
(MR). creation of a 0.5- to l-cm "safety margin" of
In our experience the use of contrast- ablated peritumoral liver tissue to destroy
enhanced ultrasound (CEUS) represents a sig- microscopic infiltrating tumor and therefore
nificant improvement over conventional US in limit the incidence of local recurrence. In
each of the above-mentioned steps and has patients with previous radical treatment for col-
proven useful to achieve optimal patient man- orectal, breast, or other primary tumors, RF
agement and treatment results (1). treatment is feasible when up to five liver

135
136 M. Tonolini and L. Solbiati

metastases are present, each one not exceeding vascular imaging to tissue perfusion imaging
4cm in size (5-10). (14).
Cross-sectional imaging modalities such as Many authors used CEUS examination in
multiphasic contrast-enhanced helical CT and the late liver-specific phase, 2 to 5 minutes
dynamic gadolinium-enhanced MR represent after the administration of an air-based first-
the mainstay for staging hepatic and extrahep- generation contrast agent such as Levovist.
atic neoplastic involvement. The use of hepato- Malignant tumors appear as hypoechoic defects
biliary and reticuloendothelial-specific MR in the brightly enhanced liver parenchyma
contrast agents may be helpful in selected achieving both an increase in lesion conspicu-
patients to maximize lesion detection (11,12). ity, particularly for subcentimeter lesions, and
The "gold standard" for detection of focal better characterization of lesions as malignant.
liver lesions undoubtedly is by intraoperative Improvement in sensitivity for the detection of
ultrasound (IOUS). Unenhanced, B-mode individual metastases was reported to be from
ultrasound is cheap, fast, and widely available; 63-71 % to 87-91 %. Still, the examination has
therefore it is commonly used for screening of limited value in the deepest portions of the liver
focal lesions in patients with chronic liver and in patients with an unsatisfactory sono-
disease or with a history of cancer. Liver metas- graphic window (15-18).
tases display an extremely varied sonographic Second-generation microbubble contrast
appearance, even within the same patient, as agents such as SonoVue (sulfur hexafluoride)
well as during the course of therapy (13). have higher harmonic emission capabilities and
The sensitivity of US depends on the opera- prolonged longevity. Their use, coupled with
tor's experience and available equipment, very low acoustic power (mechanical index
acoustic window from the patient's body values 0.1-0.2) that limits microbubble destruc-
habitus, bowel gas distention, and enlargement tion, allows a continuous-mode CEUS exami-
and inhomogeneity of the liver parenchyma nation. Signals from stationary tissues are
due to steatosis, fibrosis, chronic liver disease canceled and only harmonic frequencies gener-
and postchemotherapeutic changes. ated by microbubbles are visualized. This
The rate of detection of hepatic metastases imaging modality enables the display of both
with unenhanced ultrasound is lower than that macro- and microcirculation. The enhancement
of CT and MR; the reported accuracy ranges is displayed in real time over the arterial,
from 63% to 85%. The sensitivity of US is early/full portal, and delayed phases. Motion
particularly poor for smaller focal lesions (less or color blooming artifacts characteristic of
than 1em). Moreover, only limited characteri- color and power Doppler sonography are not
zation of focal liver lesions is possible with present. Although SonoVue has no live-specific
baseline US (13). To overcome the limitations accumulation, significant enhancement persists
of conventional US and increase the detection in the liver parenchyma 4 to 5 minutes after the
of focal lesions for accurate disease staging, injection (1).
CEUS has proved to be a valuable tool (1,13). In our experience continuous-mode CEUS
The recent development of contrast-specific can significantly improve the accuracy of ultra-
ultrasound software systems by the major sound for the detection, characterization, and
ultrasound companies (systems for pulse inver- staging of liver tumors, reaching very high sen-
sion, phase inversion, contrast-tuned imag- sitivity for the detection of both hyper- and
ing, coherent contrast imaging, contrast pulse hypovascular liver malignancies. In particular,
sequencing, etc.), all based on the principle of in cancer patients increased conspicuity for
wideband harmonic sonography that displays small hypovascular metastases with detection
microbubble enhancement in gray scale with of satellite and additional previously invisible
optimal contrast and spatial resolution, facili- lesions, leads to a modification in the therapeu-
tated the evaluation of the microcirculation. tic approach with exclusion of treatment in
This prompted the evolution of CEUS from 25% of patients.
11. Ultrasound Imaging in Tumor Ablation 137

Given these capabilities of CEUS, before 1. Small HCCs detected by CT/MR but
performing ablative therapies in our institution not visible with unenhanced US against an
our diagnostic workup includes laboratory tests inhomogeneous cirrhotic liver parenchyma;
and tumor markers along with conventional these lesions may be reached only during
and contrast-enhanced US. At least one cross- the transient arterial phase of CEUS, in which
sectional imaging modality (CT and/or MRI) they appear as hyperenhancing foci (19)
performed no more than 1 week prior to the (Fig. 11.1);
therapeutic session also is obtained. 2. Small, usually subcentimeter hypovascu-
lar metastases, barely or not perceptible with
unenhanced US, but clearly evident as focal
hypoenhancing nodules in the portal or late
Targeting of Lesions and phase of CEUS (Fig. 11.2);
3. Areas of residual untreated or locally
Guidance of RF Ablation recurrent tumor, both primary and metastatic;
Treatment unenhanced US can almost never differentiate
between coagulation necrosis and viable tumor,
In consideration of its advantages, including but the difference is usually straightforward
nearly universal availability, portability, ease during CEUS in which viable tumor displays
of use, and low cost, US represents the mod- its native, characteristic enhancement pattern
ality of choice for the guidance of percu- and coagulation necrosis is avascular (see Fig.
taneous interventional procedures. Quick and llAA-C).
convenient real-time visualization of elec-
trode positioning is a characteristic feature
of sonography, whereas the same proce- Assessment of Treatment Results
dure may be cumbersome in CT and MR
environments. Local treatment of neoplastic liver lesions with
In our institution pretreatment CEUS the application of RF ablation induces the for-
examination is repeated as the initial step of the mation of coagulation necrosis. Obtaining ade-
RF ablation session during the induction of quate necrosis, and therefore effective tumor
anesthesia, to reproduce mapping of lesions as eradication, may be limited by the inhomo-
shown on CT/MRI examinations. Images or geneity of heat deposition and by the cooling
movie clips are again digitally stored to be com- effect of blood flow (20).
pared with immediate postablation study. During the treatment, a progressively in-
Continuous-mode CEUS allows real- creasing hyperechogenic "cloud" that corre-
time targeting of lesions, which means precise sponds to gas microbubble formation and tissue
needle insertion performed during the spe- vaporization appears around the distal probe
cific phase of maximum lesion conspicuity. In and may persist for some minutes. B-mode
the arterial phase for highly vascularized sonographic findings observed during and after
lesions such as hepatocellular carcinoma RF energy application (mostly the diameter of
(HCC) and hypervascular metastases, and in the hyperechoic region) represent only a rough
the portal or equilibrium phases for hypovas- estimate of the extent of induced coagulation
cular lesions such as colorectal and breast necrosis and therefore are not useful to reliably
cancer metastases is when needle insertion is assess treatment completeness (20,21). Simi-
optimally performed. larly, color-flow and power Doppler US are
Real-time guidance of needle positioning unreliable to evaluate the adequacy of ther-
during CEUS does not significantly prolong the apy of HCCs treated with RF ablation.
total duration of the RF ablation session. In our Furthermore, additional repositioning of elec-
opinion, this approach is considered mandatory trodes may be hindered by the hyperechoic
for the following indications: focus.
138 M. Tonolini and L. Solbiati

A B

c D
FIGURE 11.1. Tiny (9mm) hepatocellular carcinoma HCC. After bolus injection of 2.4mL of SonoVue,
(HCC) detected with contrast-enhanced helical com- the lesion is clearly visible (arrow) (C) and the cool
puted tomography (CT) in arterial phase at segment tip needle for RF ablation (Radionics, Burlington,
4 (A), in patient with additional two larger HCCs VT) is inserted into the target during the arterial
in the right lobe. During the same ablation session phase of enhanced sonography (D) with clear real-
all three HCCs are scheduled for treatment, but B- time control of the procedure.
mode sonography (B) does not detect the smallest

The assessment of the size of the induced studies (22). Imaging features on immediate
coagulation necrosis and therefore of the posttreatment CT/MR examinations, along
completeness of the tumor ablation usually is with patterns of complete and partial necrosis,
accomplished for both hepatoma and metasta- are discussed elsewhere in this book.
tic lesions by means of contrast-enhanced Most reported series demonstrate a high
helical CT or MRI. Being practically unfeasible rate of apparently complete tumor necrosis on
when ablations are performed in the interven- initial postablation evaluation. However, local
tional US room, biphasic CT (or less frequently recurrences occur frequently and result from a
dynamic MRI) is performed on the first day or lack of complete ablation (19,23). Incomplete
within 1 week after the RF ablation session and tumor necrosis determines the need for addi-
compared with baseline examinations to differ- tional treatment sessions with more patient dis-
entiate between treated regions and residual comfort and increased costs.
viable tumor that requires additional treatment Furthermore, delayed retreatment often is
(20). Findings with both modalities can pre- technically difficult owing to the unreliable
dict the extent of coagulation area to within 2 discrimination of active tumor from coagula-
to 3 mm, as a radiologic-pathologic correla- tion necrosis with unenhanced sonography
tion demonstrated in experimental and clinical against an inhomogeneous liver parenchyma
11. Ultrasound Imaging in Tumor Ablation 139

c
FIGURE 11.2. An 1.8-cm liver metastases from colo- spondence. However, the metastasis is not seen with
rectal carcinoma at segment 8. The ablation proce- B-mode sonography (A). (B) After bolus adminis-
dure is performed using a multimodality navigator tration of 2.4 mL of SonoVue, the lesion is clearly
system, which allows the simultaneous visualiza- demarcated as a hypovascular area, corresponding to
tion of helical CT (performed earlier) and B-mode the CT pattern (on the right). This allows targeting
sonography. Both real-time ultrasound (US) and CT the lesion with the cool-tip electrode in real time
move simultaneously with excellent spatial corre- during the portal enhancement phase (C) (arrow).
140 M. Tonolini and L. Solbiati

because of chronic liver disease, steatosis, and completion of the RF session with the patient
presence of previous treated areas. Effective still under general anesthesia. We use a second-
targeting of residual tumor foci is often impos- generation contrast agent (SonoVue; Bracco;
sible and re-treatment has a higher rate of Milan, Italy) (Figs. 11.3 and 11.4). Comparison
failure (19,24). of immediate postablation images with stored
Therefore, imaging strategies that enable preablation scans is essential. As visible on
rapid assessment of the extent of tissue destruc- contrast-enhanced CT and MRI, a thin and
tion induced by thermal ablation are desirable. uniform enhancing rim corresponding to reac-
Since the ablative treatment leads to the dis- tive hyperemia may surround the periphery of
ruption of tumor vascularity, the demon- the necrotic area.
stration of disappearance of any previously Residual viable tumor, usually located at the
visualized vascular enhancement inside and at periphery of a lesion, maintains the enhance-
the periphery of the tumor by means of con- ment behavior characteristic of native lesions
trast-enhanced imaging methods is the hall- visualized on pretreatment studies. Partial
mark of complete treatment of a focal liver necrosis of HCC may be diagnosed when a
tumor (25). portion of the original lesion still has hypervas-
Since the introduction of first-generation cular enhancement in the arterial phase (Fig.
ultrasound contrast agents, the use of micro- 11.5). Residual untreated hypovascular metas-
bubbles allowed better depiction of microcir- tases sometimes appear indistinguishable from
culation and parenchymal blood flow compared necrosis in the portal and equilibrium phases;
with conventional color and power Doppler. with CEUS, evaluation of the early phase is
Initial experiences with enhanced color and important since viable tumor shows weak but
power Doppler imaging addressed the evalua- perceptible enhancement (25).
tion of response of HCC to interventional If even questionable residual tumor foci with
treatments including RF ablation. Better enhancement or vascular supply are depicted,
differentiation between perfused and nonper- we perform immediate CEUS-guided targeted
fused tissue and accurate detection of persis- re-treatment. The treatment session ends only
tent viable HCC after ablation, compared with when complete avascularity is demonstrated. In
conventional color and power Doppler has our experience this approach greatly simplifies
been reported (26,27). Similarly, our group patient management and reduces costs by
demonstrated that enhanced color/power decreasing both the number of RF procedures
Doppler sonography could detect residual and follow-up examinations.
tumor immediately after RF ablation of liver In the study period 2000 to 2002 at our insti-
metastases, prompting repeat treatment ses- tution no residual tumor was detected with
sions in some cases (28). CEUS in 176 of 199 liver malignancies treated;
More recently, CEUS has been employed to of these, CT depicted residual foci in only four
reveal residual enhancement after ablation. cases (specificity 97.7%) all of which were very
CEUS has demonstrated agreement and com- small (0.8-1.7cm). In the remaining 23 tumors,
parable accuracy to helical CT, the latter single or multiple (1.0-2.2cm) residual viable
adopted as the "gold standard" after different tumor portions were visible and immedi-
treatment modalities (RF, PEl, TACE) for ately submitted to additional RF application
HCC (29,30). Meloni et al (31) reported an in the same session until no further residual
increase in sensitivity from 9.3% with contrast- enhancement was detectable. In only two cases
enhanced power Doppler to 23.3% with pulse a 1.2- to 1.9-cm residual tumor was demon-
inversion CEUS for the detection of residual strated by CT. The routine adoption of CEUS
HCC after RF ablation. (as the only technical improvement) achieved a
In our protocol, immediate postablation final result of a decrease of partial necrosis
evaluation using continuous-mode CEUS is from 16.1 % to a 5.1 % rate in 429 hepatocellu-
performed 5 to 10 minutes after the assumed lar and metastatic treated lesions.
11. Ultrasound Imaging in Tumor Ablation 141

A c

B o

FIGURE 11.3. Hepatocellular carcinoma at segment


4 in a cirrhotic patient. The 3.5-cm lesion is poorly
visualized with B-mode sonography (A), but
clearly detected with contrast-enhanced ultrasound
(CEUS) (B) and contrast-enhanced CT (C) as a
hypervascular tumor. Ten minutes after completing
ablation, CEUS is repeated and shows a hypovascu-
lar area of coagulative necrosis (D) with peripheral
extralesional hyperemic halo due to postablative
hyperemia. The size of the necrotic area is larger
than the original HCC, suggesting complete abla-
tion. (E) Contrast-enhanced CT confirms the result
of CEUS (complete ablation). E
142 M. Tonolini and L. Solbiati

A B

FIGURE 11.4.
11. Ultrasound Imaging in Tumor Ablation 143

G H

FIGURE 11.4. (continued) In a patient treated with Under real-time guidance with enhanced sonogra-
percutaneous cool-tip RF ablation 2 years ago for a phy the recurrence is treated with cool-tip needle RF
large colorectal metastases at segment 8, the serum ablation (arrow) with gas formation in the area of
level of carcinoembryonic antigen (CEA) increased. treatment (D). Seven minutes after removing the
B-mode unenhanced sonography shows a large mass RF electrode, a second study with SonoVue is
at segment 8, but cannot differentiate the necrotic performed and the recurrence appears avascular in
area from previous ablation from local recurrence both arterial (E) and portal (F) phase, suggesting
(A). After bolus injection of SonoVue, the larger complete treatment. The excellent correlation of
lateral portion of the mass remains avascular in both enhanced sonography and contrast-enhanced helical
arterial (B) and portal (C) phase, while the smaller CT is clearly appreciable; a previous necrotic area
medial portion (arrows) appears hypervascular in and local recurrence are shown in the arterial and
arterial phase (B) and hypovascular in portal phase portal phases before ablation (G,H) and at 24 hours
(C) and therefore corresponds to local recurrence. following ablation (I,J).
144 M. Tonolini and L. Solbiati

c D
FIGURE 11.5. Contrast-enhanced sonography per- repeated 6 minutes after withdrawing the electrode
formed during the ablation session can help to and clearly shows that only a small central portion
achieve complete treatment and thus avoid the need of the mass is actually avascular (E). Therefore,
for local re-treatment. A large (5-cm) HCC is seen immediate new insertion of an RF electrode is per-
in the right lobe before ablation with B-mode sonog- formed under the guidance of enhanced sonography.
raphy (A) and SonoVue-enhanced sonography in As a result, complete devascularization of the
arterial phase (B). After precise targeting with RF mass is demonstrated with further administration of
electrode (arrow) (C), apparently optimal treatment microbubbles at the end of ablation (F) and con-
is performed, with formation of a wide diffusion of firmed with contrast-enhanced helical cr in arterial
gas (D). However, SonoVue-enhanced sonography is phase at 24 hours (G).
11. Ultrasound Imaging in Tumor Ablation 145

E F

FIGURE 11.5. (continued)

Long-Term Follow-Up mode CEUS has proved to be of value to


confirm or exclude possible or suspicious local
Contrast-enhanced helical CT and, in some in- recurrences and metachronous new lesions
stitutions, dynamic gadolinium-enhanced MRI detected by cross-sectional imaging and to
are the mainstays for imaging follow-up of assess the possibility of their CEUS-guided
treated patients and the detection of local, retreatment.
remote intrahepatic and extrahepatic disease
relapse (32-35). Recently, the use of functional
imaging with fluorine-18-fluoro-2-deoXY-D- References
glucose (FDG) proved superior to cross-
1. Solbiati L, Tonolini M, Cova L, Goldberg SN.
sectional studies (36). The role of contrast-enhanced ultrasound in the
At our institution serum tumor markers are detection of focal liver lesions. Eur Radiol 2001;
correlated with biphasic helical CT obtained 11(SuppI3):EI5-E26.
every 3 to 4 months in the long-term follow- 2. Livraghi T, Goldberg SN, Lazzaroni S, Meloni F,
up of patients treated for both primary and Solbiati L, Gazelle GS. Small hepatocellular car-
metastatic liver malignancies. Continuous- cinoma: treatment with radiofrequency ablation
146 M. Tonolini and L. Solbiati

versus ethanol injection. Radiology 1999;210: 16. Albrecht T, Hoffmann CW, Schmitz SA, et al.
655-661. Phase-inversion sonography during the liver
3. Livraghi T, Goldberg SN, Lazzaroni S, et al. specific late phase of contrast enhancement:
Hepatocellular carcinoma: radiofrequency abla- improved detection of liver metastases. AJR
tion of medium and large lesions. Radiology 2001;176:1191-1198.
2000;214:761-768. 17. Quaia E, Bertolotto M, Forgacs B, et al. Detec-
4. Buscarini L, Buscarini E, Di Stasi M, Vallisa D, tion of liver metastases by pulse inversion
Quaretti P, Rocca A. Percutaneous radiofre- harmonic imaging during Levovist late phase:
quency ablation of small hepatocellular carci- comparison with conventional ultrasound and
noma: long-term results. Eur Radiol 2001;11: helical CT in 160 patients. Eur Radiol 2003;13:
914-992. 475-483.
5. Solbiati L, Ierace T, Goldberg SN, et al. Percuta- 18. Albrecht T, Blomley MJK, Burns PN, et al.
neous US-guided radiofrequency tissue ablation Improved detection of hepatic metastases with
of liver metastases: treatment and follow-up in pulse-inversion US during the liver-specific
16 patients. Radiology 1997;202:195-203. phase of SHU 508A: multicenter study. Radiol-
6. Solbiati L, Goldberg SN, Ierace T, et al. Hepatic ogy 2003;227:361-370.
metastases: percutaneous radio-frequency abla- 19. Numata K, Isozaki T, Ozawa Y, et al. Percuta-
tion with cooled-tip electrodes. Radiology 1997; neous ablation therapy guided by contrast-
205:367-373. enhanced sonography for patients with
7. Solbiati L, Livraghi T, Goldberg SN, et al. Per- hepatocellular carcinoma. AJR 2003;180:143-
cutaneous radiofrequency ablation of hepatic 149.
metastases from colorectal cancer: long-term 20. Goldberg SN, Gazelle GS, Mueller PRo Thermal
results in 117 patients. Radiology 2001;221: ablation therapy for focal malignancy. A unified
159-166. approach to underlying principles, techniques,
8. Livraghi T, Goldberg SN, Solbiati L, Meloni F, and diagnostic imaging guidance. AJR 2000;174:
Ierace T, Gazelle GS. Percutaneous radio- 323-331.
frequency ablation of liver metastases from 21. Leyendecker JR, Dodd GD, Halff GA, et al.
breast cancer: initial experience in 24 patients. Sonographically observed echogenic response
Radiology 2001;220:145-149. during intraoperative radiofrequency ablation
9. De Baere T, Elias D, Dromain C, et al. Radiofre- of cirrhotic livers: pathologic correlation. AJR
quency ablation of 100 hepatic metastases with 2002;178:1147-1151.
a mean follow-up of more than 1 year. AJR 2000; 22. Goldberg SN, Gazelle GS, Compton CC, Mueller
175:1619. PR, Tanabe KK. Treatment of intrahepatic
10. Solbiati L, Ierace T, Tonolini M, et al. Radiofre- malignancy with radiofrequency ablation: radio-
quency thermal ablation of hepatic metastases. logic-pathologic correlation. Cancer 2000;88:
Eur J Ultrasound 2001;13:149-158. 2452-2463.
11. Sica GT, Ji H, Ros PRo CT and MR imaging of 23. Chopra S, Dodd GD III, Chintapalli KN,
hepatic metastases. AJR 2000;174:691-698. Leyendecker JR, Karahan 01, Rhim H. Tumor
12. Valls C, Andia E, Sanchez A, et al. Hepatic recurrence after radiofrequency thermal abla-
metastases from colorectal cancer: preoperative tion of hepatic tumors: spectrum of findings on
detection and assessment of resectability with dual-phase contrast-enhanced CT. AJR 2001;
helical CT. Radiology 2001;218:55-60. 177:381-387.
13. Harvey CJ, Albrecht T. Ultrasound of focal liver 24. De Baere T, Elias D, Dromain C, et al. Radio-
lesions. Eur RadioI2001;11:1578-1593. frequency ablation of 100 hepatic metastases
14. Lencioni R, Cioni D, Bartolozzi C. Tissue har- with a mean follow-up of more than 1 year. AJR
monic and contrast-specific imaging: back to 2000;175(6):1619-1625.
gray scale in ultrasound. Eur Radiol 2002;12: 25. Rhim H, Goldberg SN, Dodd GD III, et al.
151-165. Essential techniques for successful radiofre-
15. Harvey CJ, Blomley MJK, Eckersley RJ, et al. quency thermal ablation of malignant hepatic
Hepatic malignancies: improved detection with tumors. Radiographies 2001;21:S17-S31.
pulse-inversion US in the late phase of enhance- 26. Choi D, Lim HK, Kim SH, et al. Hepatocellular
ment with SHU 508A-early experience. Radi- carcinoma treated with percutaneous radio-
ology 2000;216:903-908. frequency ablation: usefulness of power Doppler
11. Ultrasound Imaging in Tumor Ablation 147

us with a microbubble contrast agent in evalu- quency ablation. Comparision of pulse inversion
ation therapeutic response-preliminary study. contrast-enhanced harmonic sonography, con-
Radiology 2000;217:558-563. trast-enhanced power Doppler sonography and
27. Cioni D, Lencioni R, Rossi S, et al. Radiofre- helical CT. AJR 2001;177:375-380.
quency thermal ablation of hepatocellular 32. Choi H, Loyer EM, DuBrow RA, et al. Radio-
carcinoma: using contrast-enhanced harmonic frequency ablation of liver tumors: assessment of
power Doppler sonography to assess treatment therapeutic response and complications. Radio-
outcome. AJR 2001;177:783-788. graphies 2001;21:S41-S54.
28. Solbiati L, Goldberg SN, Ierace T, Della Noce M, 33. Catalano 0, Lobianco R, Esposito M, et al.
Livraghi T, Gazelle GS. Radio-frequency Hepatocellular carcinoma recurrence after per-
ablation of hepatic metastases: postprocedural cutaneous ablation therapy: helical CT patterns.
assessment with a US microbubble contrast Abdom Imaging 2001;26:375-383.
agent-early experience. Radiology 1999;211: 34. Dromain C, De Baere T, Elias D, et al. Hepatic
643-649. tumors treated with percutaneous radio-
29. Ding H, Kudo M, Onda H, et al. Evaluation of frequency ablation: CT and MR imaging follow-
posttreatment response of hepatocellular carci- up. Radiology 2002;223:255-262.
noma with contrast-enhanced coded phase- 35. Sironi S, Livraghi T, Meloni F, et al. Small hepa-
inversion harmonic US. Comparison with tocellular carcinoma treated with percutaneous
dynamic CT. Radiology 2001;221(3):712-730. RF ablation: MR imaging follow-up. AJR 1999;
30. Numata K, Tanaka K, Kiba T, et al. Using con- 173:1225-1229.
trast-enhanced sonography to assess the effec- 36. Anderson GS, Brinkmann F, Soulen MC, et al.
tiveness of transcatheter arterial embolization FDG position emission tomography in the
for hepatocellular carcinoma. AJR 2001;176: surveillance of hepatic tumors treated with
1199-1205. radiofrequency ablation. Clin Nucl Med 2003;28:
31. Meloni MF, Goldberg SN, Livraghi T, et al. Hepa- 192-197.
tocellular carcinoma treated with radiofre-
12
Magnetic Resonance Imaging
Guidance for Tumor Ablation
Koenraad 1. Mortele, Stuart G. Silverman, Vito Cantisani, Kemal Tuncali,
Sridhar Shankar, and Eric vanSonnenberg

Since the late 1980s, magnetic resonance the abdomen, pelvis, breast, and musculoskele-
imaging (MRI) has been added to ultrasound tal system.
(US) and computed tomography (CT) as a
cross-sectional imaging tool that can be used
to guide the interventional diagnosis and Interventional Guidance:
treatment of a variety of disorders. Due to its
superior soft tissue contrast, multiplanar capa-
Modality Comparison
bilities, lack of ionizing radiation, and, most
importantly, ability to image tissue function and
Role of Imaging in Image-Guided
temperatures, MRI has been suggested as the
Therapy
ideal tool to guide minimally invasive therapies All image-guided percutaneous tumor abla-
(1,2). Nevertheless, when the concept of percu- tions require five processes: (a) planning, (b)
taneous MRI-guided tumor ablation was first targeting, (c) monitoring, (d) controlling, and
introduced, restricted patient access and (e) assessing. Planning is done before the pro-
limited spatial and temporal resolution with cedure and determines whether the interven-
conventional high- and mid-field MRI systems tion is appropriate, and how the lesion can be
limited its widespread use. Technical advances best approached. This also includes a decision
in both open-configuration magnet design and regarding which image guidance modality best
the development of fast gradient-echo pulse accomplishes the requirements for this task.
sequences have contributed substantially to an The first step in the procedure is targeting the
increasing interest in MRI-guided interven- lesion, typically with a biopsy needle or therapy
tions (1,2). As a consequence, percutaneous probe. For this, accurate depiction of the tumor
tumor ablations now can be performed under and surrounding anatomy is critical. All of the
real-time MRI guidance. instruments used to perform the procedure
This chapter discusses the rationale for using should be visible so that adjustments can be
MRI to plan, guide, monitor, and control per- made as the lesion is targeted. Real-time, inter-
cutaneous tumor ablations; reviews the history active, and multiplanar imaging are helpful in
and development of open-configuration MRI targeting. For example, real-time imaging
systems and how they can be used to guide per- allows the operator to obtain instantaneous
cutaneous interventions; highlights the unique feedback of image information that can be used
principles and issues of designing and imple- to make adjustments. A procedure that is inter-
menting an interventional MRI suite; and active allows the operator to make adjustments
discusses the current status of MRI-guided in the course or trajectory of the needle and
tumor ablation trials that use a variety of abla- immediately assess the effects of those adjust-
tive agents in several organ systems, including ments. Visualizing the tumor and instrument in

148
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 149

multiple planes gives the operator a three- allow direct visualization of the entire needle
dimensional (3D) understanding of the target. tract in one image (3). Although US can be used
Being able to monitor the ablation and the to image in multiple planes, the path of the
tissue changes are other critical components of needle tract may be obscured by bony struc-
image-guided ablation. Monitoring should tures such as the ribs or air from interposed
depict the location and extent of ablative bowel loops (3).
effects and allow the operator to differentiate Magnetic resonance imaging-guided inter-
treated from untreated tumor, and affected ventions depend on the ability of the interven-
from unaffected normal tissue. Similarly, the tional radiologist to position a needle probe
ability to control the changes induced by the under MRI guidance into a relatively small
procedure or the ability to change the amount target within the body. Therefore, research and
and location of the ablation intraprocedurally development of MRI-guided biopsy techniques
is important. Monitoring and controlling are have been an important and fundamental
essential components of image guidance aspect of the beginning of interventional MR.
because it is not always possible to predict accu- Mueller et al (4) first described the use of an
rately the size and shape of an ablative lesion. MR-compatible needle and showed that after
Only with imaging can an ablation protocol be US-guided placement, both the liver lesion and
optimized to cover a tumor maximally, while the needle could be viewed using a conven-
minimizing effects in normal tissues. Finally, fol- tional MRI system. Silverman et al (5) reported
lowing the procedure, imaging is used to assess the first MRI-guided biopsy wherein the
the extent of ablative changes, the complete- entire procedure was performed under MRI
ness of the ablation, and complications. guidance. This technique employed a 0.5-T
vertically open configuration MRI system
specifically designed to guide radiologic inter-
Image-Guided Ablations: Magnetic vention and surgery (Signa SP; GE Medical
Resonance Imaging Advantages Systems; Milwaukee, WI). In addition to being
Given these five critical components of image- open, the system contained two important fea-
guided ablations, each guidance modality has tures: (a) images were displayed to the opera-
inherent benefits and disadvantages. Planning tor inside the room on two MR-compatible
requires visualization of the lesion and the sur- monitors mounted above the field, that allowed
rounding anatomy, and a survey of the rest of ongoing review of images during procedures;
the target organ for additional lesions. In (b) the system contained an integrated optical
clinical practice, MRI is often not needed localizer and tracker, which allowed interactive
because US and CT demonstrate most lesions. control of the biopsy needle. Subsequently,
However, MRI is a sensitive technique for Frahm et al (6) and Lu et al (7) each reported
imaging and can be helpful in surveying for MRI-guided biopsy using a 0.2-T horizontally
other sites of disease. Ultrasound and CT are open configuration MRI system in which free-
commonly used for targeting also, but MRI- hand techniques using fast gradient-echo
guidance may be indicated in patients with sequences were utilized. Fast gradient-echo
lesions that either are not visualized by US and sequences were used successfully to guide liver
CT (e.g., breast and prostate cancers), or are biopsies in closed magnet systems (8,9).
visible by CT and MRI but not with US and Similarly, although transrectal ultrasound
require angling the needle, such as a liver lesion (TRUS) is useful in the vast majority of
beneath the hepatic dome or an adrenal gland prostate gland biopsies, MRI may be used in
mass (Fig. 12.1). In such cases, using CT guid- patients: in whom the serum prostate specific
ance, if the pleural space and lung are avoided, antigen (PSA) is elevated and rising despite a
lesions in the upper abdomen can be reached negative TRUS biopsy, in whom the lesion is
by either angling the gantry or angling the visible only with MRI imaging, or in whom the
needle using the triangulation method. The tri- rectum is absent (e.g., post-abdominoperineal
angulation method is limited in that it does not resection) and thus TRUS-guided biopsy is not
150 K.J. Mortele et al

A B

c D

FIGURE 12.1. Magnetic resonance imaging (MRI)-guided


percutaneous biopsy of hepatic metastasis of gastrointesti-
nal stromal tumor (GIST). (A-C) Axial, sagittal, and
coronal gradient-echo Tl-weighted images obtained with
the open MR scanner during the procedure planning show
the expected trajectory of the needle using the optical
tracking system. The lesion (arrow) is located in segment 8
beneath the diaphragm. (D,E) Oblique sagittal and coronal
gradient-echo T1-weighted images show the presence of a
E fine biopsy needle (22-gauge) at the edge of the mass.
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 151

There are many ways in which an interven-


tional device can be tracked during a procedure
using MR guidance. Most procedures reported
to date are performed using passive tracking in
which imaging is used to follow the needle. In
passive tracking, the needle or probe is visual-
ized by the signal void created by the needle's
magnetic susceptibility artifact. The needle can
be seen as long as the imaging plane includes
it. The frequency with which the needle is seen
is determined by how fast the images are
acquired, reconstructed, and shown to the oper-
ator during the procedure (15,16). Some open
interventional MR systems, by using an optical
tracking system, automatically image the device
(e.g., a needle) because it is attached to a probe
whose position is sensed by the optical tracker.
FIGURE 12.2. MRI-guided percutaneous transgluteal As the probe is moved, its position is sensed,
biopsy of adenocarcinoma in the prostate gland in a and the scan plane is changed. Other methods
patient post-abdominoperineal resection for rectal of localization include active tip tracking,
cancer. Axial TSE T2-weighted image obtained with mechanical sensors, and radiofrequency (RF)
the open MR scanner during the procedure shows localizers (17-20). Active tip-tracking is
two biopsy needles targeting the low signal intensity achieved by embedding a miniature RF coil in
areas (arrows) in the apex of the prostate. the tip of a needle or therapy probe. This allows
active intraprocedural identification of the
possible. The first group is quite numerous and spatial position of the tip. The most important
includes anxious men who are facing rising advantages of active tracking over passive
PSA levels with no diagnosis. Magnetic reso- tracking are its high temporal resolution and
nance imaging-guided prostate gland biopsy the option to use the three-dimensional coor-
may be done either in a closed bore system dinates to steer the scan plane (17-20).
using a transgluteal approach, or in an open Although MRI can be used to plan an abla-
system using a transrectal or transperineal tion and target a tumor, its ability to monitor
approach (Fig. 12.2) (10). (and thus control) ablations is its greatest
Because studies have shown that MRI is both advantage (21). Through MR monitoring,
sensitive and specific for the detection of breast thermal lesion size and configuration can be
cancer, MRI has been used to biopsy the breast directly controlled by the interventionalist and
(11,12); MRI is needed because many lesions adjusted during the procedure. There has been
are not seen at x-ray mammography. Targeting intense interest in developing noninvasive
is achieved either using template guidance at methods of monitoring temperature distribu-
high-field closed systems (13) or freehand, with tions in vivo, since the biologic efficacy of
open systems (14). Although interventions are thermal-ablation techniques is strongly depen-
feasible with closed MRI systems, they can be dent on achieving desired temperatures in all
cumbersome as the patient needs to be moved parts of the tumor. Temperature-sensitive MR
out ofthe magnet bore to manipulate the instru- sequences have been developed to enable
ments, and, therefore, the procedure is only accurate online monitoring of heat deposition
intermittently imaged. As a consequence, since (22-24). Three-dimensional mapping of tem-
most structures move with respiration, targeting perature changes with MRI are based on
may be difficult. Generally, open systems offer relaxation time T1, diffusion coefficient (D), or
improved patient access during the procedure proton resonance frequency (PRF) of tissue
compared with closed systems. water. Temperature-sensitive contrast agents
152 K.J. Mortele et al

also may be used (25,26). Among these strength. The gradient performance of these
methods of MRI-based temperature mapping, systems is good and ranges typically between 10
the PRF-based method is the preferred choice and 15 mT/m, providing sufficient image quality
for many applications at mid- and high field for guiding tumor ablations (30-32).
(~1 T), because of its sensitivity and linear rela-
tion with temperature, and near-independence Closed Systems
with respect to tissue type. The PRF methods
utilize RF-spoiled gradient-echo imaging Interventional MRI-guided procedures also
methods to measure the phase change in may be performed using conventional MR sys-
resonance that results from the temperature- tems (22). Closed systems, operating at higher
dependent change in resonance frequency. field strengths, provide better image quality
When field homogeneity is poor due to small by means of a higher signal-to-noise ratio.
ferromagnetic parts in the needle, or at low However, patient access is limited and instru-
field strengths (::;;0.5T), diffusion and Tl- ment manipulations are performed when the
based methods may provide better results. patient is outside the magnet, or when using a
Temperature-sensitive contrast agents may be system with a flared opening, with the operator
particularly suitable when absolute thermo- leaning in. Some lesions, however, may be visu-
regulation is desired, for example, when an alized only using a conventional closed MRI
indication exists for exceeding a certain tem- system and not with an open MRI system of
perature threshold (26). lower field strength. MRI-guided procedures in
both closed and open systems may not be pos-
sible in large patients because the space within
the magnet bore typically is restricted.
History and Development of
Interventional MRI Interventional MRI Suites
Interventional MRI Systems An MR-compatible environment is needed to
support an interventional MR scanner (2).
Open Systems
Important principles of installing an interven-
In the mid- to late 1980s, open-configuration tional MR scanner relate to issues of siting,
MRI systems were developed primarily for the patient care, and MR-compatible instrumenta-
purpose of combating claustrophobia and tion (1). Siting an interventional MR scanner
aiding the MRI of pediatric patients. Forward- must take into account essential issues related
thinking interventional radiologists, however, to MRI scanners: magnetic shielding, RF shield-
used these open MRI systems to gain access to ing, floor loading, vibration, acoustic dampen-
patients to perform a variety of interventions ing, and cryogen venting and storage. Factors
(27-29). As a consequence, interventional MRI related to siting the scanner in an interventional
systems were developed that provided guid- suite or operating room also need to be consid-
ance systems integrated with open configura- ered: control of air qualitylflow, humidity, tem-
tion MRI systems that allowed intervention perature, zoned lighting, the inclusion of scrub
and surgery to occur simultaneously with sinks, restricted and semirestricted areas, and
imaging (28,29). Open-configuration MRI scan- ports for suction, gases, and therapy devices.
ners are now produced by most major MRI Issues related to patient care include the
manufacturers. In general, access to the patient ability of all operators to have access to the
may be achieved via a horizontally open system patient for the purpose of performing the pro-
(of which there are many-Magnetom Open cedure, observing and monitoring the patient,
System; Siemens Medical Solutions; Erlangen, and administering drugs and/or anesthesia.
Germany, is one) or a vertically open system Emergency therapy must be able to be per-
(e.g., Signa SP; General Electric Medical formed. Emergency treatment can be per-
Systems; Milwaukee, WI). Most of these formed inside the procedure room with
systems are either low or midfield in magnet MR-compatible devices, such as airways and
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 153

intravenous tubing. Cardiac defibrillators are MRI Compatible Instruments


not MR-compatible. Therefore, to treat ventric-
ular fibrillation, either the magnet needs to be All instruments used in the interventional MR
quenched or the patient brought out of the procedure room must be MR-compatible. A
room. device is MR compatible if it can be used in the
Another important point is that silent scanner room during the procedure safely and
myocardial ischemia cannot be detected in an without adversely affecting the device itself or
MR environment (33,34). Therefore, before the procedure for which it is being used (1,2).
undergoing interventional MRI procedures, Magnetic resonance compatibility includes
all patients need to be screened for coronary issues related to ferromagnetism, electric inter-
artery disease and a risk/benefit assessment ference, image distortion, and device heating
made as to whether the procedure should be from RF energy (Table 12.1). Interventional
performed using MRI guidance. Magnetic and surgical instruments may cause artifacts,
resonance imaging-guided procedures should which lead to image distortion. The size of the
be performed only in patients for whom the artifacts depends mainly on the following
benefit of performing the procedure is greater factors: the size of the instrument, the orienta-
than the risk of not detecting silent myocardial tion of the instrument to the main magnetic
ischemia. Just as patients need to be screened field, the imaging sequence used, and the ori-
before undergoing an MRI examination, physi- entation of the frequency-encoding gradient
cians also need to be screened in the same (35). The amount of artifact in an MR image
fashion. The interventional MR room needs to increases with instrument size and increasing
be surrounded by a checkpoint at the 5-gauss angle of the instrument to the main magnetic
line, which is typically, depending on the field field Bo (1).
system and parameters of the system, 1 m out- Once a major obstacle, MR-compatible
side the procedure room. instrumentation now is developed such that a

TABLE 12.1. Overview of magnetic resonance (MR) compatibility.


Factor Effect Example Solution
Ferromagnetic Object moves/rotates to Most conventional surgical Change instrument materials
attraction align with magnet; may instruments
endanger occupants

Electrical interference
Device on MR Low signal-to-noise ratio. Almost anything with Provide RF shielding device;
scanner stripes. zippers, bright spots electrical plug, ferromagnetic, remove the electronics
and an interference hazard to remote location
MR scanner on Magnetic field/RF interferes Computer discs, video Provide remote electronics;
device with device often with displays use liquid crystal displays
subtle effects

Image distortion Image artifacts, spatial Signal void around needle Move devices out of image
by devices distortions field of view; match materials
to tissue susceptibility; break
up eddy current paths

Device heating Patient burns from RF Pulse oximeter wire, ECG Use nonconductive materials
from RF energy inductive heating of device wire (e.g., fiberoptics); keep
(in or out of and from contact with antennae length to a minimum;
patient) patient inspect for patient/device
grounding paths

ECG. electrocardiogram; RF, radiofrequency.


154 K.J. Mortele et al

variety of procedures can be performed (1). performed percutaneously, without the use of
An array of basic interventional and surgical surgical techniques and therefore the term
instruments including needles, scissors, eleva- cryotherapy is more appropriate. Cryoablation
tots, forceps, and blades have been manufac- and cryodestruction refer to processes that
tured for use during both interventions and occur during cryotherapy.
surgery (36). MR-compatible monitors and Cryotherapy is one of the oldest methods of
anesthesia delivery systems are used to monitor tissue destruction known to mankind. It was
patients safely. Therapy delivery systems such used even during the times of Hippocrates to
as electrocautery, laser, RF, and cryotherapy, control hemorrhage. In the 20th century and
discussed in further detail below, also are avail- currently, cryotherapy has been used for a
able for use during MRI-guided interventions. variety of cancer treatments in the brain, head
and neck, breast, and more recently, in the liver
and prostate gland. Cryotherapy destroys tissue
Percutaneous MRI-Guided mainly by cellular dehydration, cell membrane
rupture, and vascular stasis. Cell death occurs
Tumor Ablation typically at -30° to -40°C, and is dependent on
several factors including tissue type, freezing
Concomitant with the development of open- and thawing rate, duration, and depth of freez-
configuration MR systems and interventional ing (49,50). Cryotherapy is a time-tested, effec-
MRI, there has been worldwide interest in the tive ablation technique. It is not dose-limited,
use of imaging to guide percutaneous ablation blood loss is minimal, and, perhaps more
of liver tumors using both vascular and direct importantly, it is amenable to imaging. Tissue
percutaneous approaches (37). Successes in the effects can be seen with US, CT, and MRI. Fur-
focal therapy of both hepatocellular carcinoma thermore, lesions produced by cryotherapy are
(HCe) and metastatic colorectal cancer to the sharply marginated and depict the zone of
liver have been the major driving force in tissue necrosis accurately (49,50).
developing image-guided percutaneous tumor The liver and prostate gland have been the
ablation. It has led to the research of percuta- most common sites in which cryotherapy has
neous tumor ablation in organs elsewhere in been used (Fig. 12.3). In the liver, it has been
the body, including the kidney, bone, soft tissue, used in patients with HCC or metastases who
breast, and lung. The rationale for percutaneous are unable to undergo hepatic resection
tumor ablation research is to develop a less (39,43). In the operating room, US is used for
invasive, repeatable, and effective method that lesion detection, for guidance of cryosurgical
will allow more patients, including nonsurgical probe placement, and monitoring of the freez-
candidates, to be treated (38). ing process (44). Ultrasound depicts the effects
Many ablative agents have been investigated. of freezing in real time and accurately demon-
In general, these agents result in tumor cell strates the zone of necrosis that is subsequently
death using either thermal-based energy (e.g., produced (45). However, US is limited by its
heat or cold) or chemical agents such as inability to image beyond the proximal edge of
ethanol. Thermal agents include lasers, RF, the iceball because the energy is reflected at
and microwaves, all of which heat tissue, and this front edge. As a result, the tumor may not
cryotherapy, which freezes tissue (39-42). be completely visualized during the treatment,
and thus it may be incompletely treated. The
relatively small surgical incisions used during
Cryotherapy minimally invasive liver surgery also may limit
Cryotherapy is defined as therapeutic tissue the ability of US to image lesions completely
destruction in situ by freezing. Once adminis- (46). Although liver cryosurgery with intraop-
tered predominantly through "open" surgical erative ultrasound guidance has been used in
means, it was known mostly by the term many centers for more than a decade, the
cryosurgery (43-48). Now, cryotherapy can be overall survival in patients with metastatic
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 155

A B

c D

FIGURE 12.3. MRI-guided cryoablation of liver metas-


tasis. (AB) Intraprocedural axial and coronal SPGR
images show location of five cryoprobes within the
lesion (arrows) located in segment 8 of the liver. (C-E)
Axial and coronal SPGR Tl-weighted MR images
obtained during cryotherapy with five cryoprobes show
enlargement of the iceball. E
156 K.J. Mortele et al

colorectal carcinoma has not been significantly bidity that increases the risks of nephrectomy,
different from that which is achieved using and conditions that warrant a nephron-sparing
conventional surgical techniques (47). Never- procedure, such as solitary kidneys, multiple
theless, cryosurgery is used for surgically un- tumors, or renal insufficiency.
resectable disease, and because more normal Percutaneous MRI guided cryoablation of
liver can be preserved than with surgical soft tissue and bone lesions has been performed
resection. as well. Most patients are poor surgical candi-
While cryotherapy has been recognized as a dates and they are referred for palliation or
useful ablative agent in the treatment of malig- local tumor or pain control. A common indica-
nant liver lesions, it was considered a "surgical tion for soft tissue cryoablation is presacral
only" option because of the necessity for large recurrence of anorectal cancer following
probes. Recently, there has been the develop- abdominoperineal resection (APR) (Fig. 12.5).
ment of cryoneedles (as small as 18-gauge Other situations in which cryoablation has
needles) that can be placed percutaneously. proven to be helpful include treatment of soft
Tacke et al (36) reported on the use of a novel tissue sarcomas, vascular malformations, and
MRI-compatible probe in animals using a metastatic lesions (52,53).
I.5-T magnet in the rabbit liver. The rationale for MRI-guided cryotherapy is
Magnetic resonance imaging-guided cryother- summarized as follows: (a) freezing is an effec-
apy holds significant promise for several tive ablative technique and can be monitored
reasons. First, the short T2 relaxation time of ice well with MRI; (b) MRI, like US, is near real
results in excellent visualization of the iceball time and multiplanar; (c) unlike US, it can
or cryolesion, which, using either Tl- or T2- depict the entire iceball and surrounding struc-
weighted sequences, is represented as a signal tures, and the iceball can be discriminated from
void (48,49). Using a variety of pulse sequences, tumor during the procedure; (d) unlike heating,
excellent contrast between the iceball and the margins of necrosis following cryoablation
tumor can be achieved in either a multiplanar are sharp and unambiguous, and the MR assess-
or a three-dimensional format. This results in ments correlate well with necrosis. Visualizing
the accurate depiction of the entire iceball and ablative changes intraprocedurally obviates 24-
its relationship in all dimensions with the hour follow-up MRI or CT. Although tumor
treated tumor (48,49). As with ultrasound, the ablation with ethanol injection, RF, and inter-
size of the iceball or cryolesion visualized with stitiallaser therapy also may be performed with
MRI correlated closely with necrosis. Silver- imaging guidance, the amount of tissue change
man et al (31) described a method for cryo- demonstrated at US and CT during these ther-
ablating tumors percutaneously under MRI apies is limited, variable, and does not accu-
guidance. Using an optical tracking system, cry- rately reflect the zone of necrosis (54).
oneedles were placed, and liver tumors were
ablated during two freeze cycles. Using repeti-
tive multiplanar Tl-weighted fast spin echo
Ethanol Ablation
(FSE) or Tl-weighted spin echo (SE) images, Ethanol was one of the first ablative agents
MRI was used to monitor progress of the treat- used percutaneously. It is relatively easy to use,
ment, whether the lesion was adequately inexpensive, and effective in ablating tissues.
covered, and to be sure vital structures were Percutaneous injection of absolute ethanol is
avoided. Consistent with the results of animal an established form of therapy for treatment of
studies, intraprocedural appearance of iceballs small HCCs in many parts of the world (42).
correlated well with postprocedural estimates Alcohol causes tissue necrosis predominantly
of necrosis (31). by cellular dehydration and vascular thrombo-
Percutaneous cryotherapy of renal tumors sis. Two characteristics of HCC make it amen-
has been performed successfully using MRI able to alcohol injection therapy (42): (a) it is a
guidance (Fig. 12.4) (51). Indications for percu- well-vascularized, soft tumor so that ethanol
taneous tumor ablation (PTA) of renal tumors easily diffuses within it; (b) it is frequently
include elderly patients, patients with comor- encapsulated, which prevents ethanol from
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 157

A 8

c o
FIGURE 12.4. MRI-guided cryoablation of renal cell polar region of the right kidney. (B-D) Intraproce-
carcinoma (Ree) in the right kidney in a patient dural oblique axial and coronal TSE T2-weighted
post left nephrectomy. (A) Axial fast spin echo images show location of three cryoprobes within the
(FSE) T2-weighted MR image shows hyperintense lesion and enlargement of the iceball.
appearance of the renal mass (arrows) in the inter-

escaping. Metastatic tumors typically are not tinguished easily from T2-hyperintense tumor
soft or encapsulated, and therefore alcohol is during treatment. Shinmoto et al (55) showed
not as useful in the treatment of metastatic liver that a water-suppressed TI-weighted rapid
disease. acquisition with relaxation enhancement
Ultrasound and CT have been used success- (RARE) sequence could be used to differenti-
fully in the guidance for alcohol and, as such, ate alcohol from long T2 water components of
MRI has not been used widely. But MRI may tumor and edema. The future of MRI-guided
be helpful in the guidance of percutaneous alcohol ablation is uncertain, and will probably
injection of alcohol into lesions that are not be dictated by the relative benefits of alcohol as
viewed by ultrasound or CT. In addition, MR an ablative agent in the treatment of liver
monitoring of ethanol diffusion may be useful tumors.
(7,55). Lu et al (7) reported on the use of MRI
in the treatment of hepatic lesions in anes-
thetized pigs. Their study showed that ethanol
Laser Ablation
appeared hypointense on T2-weighted images. Bown (56) described interstitial hyperthermia
They postulated that if ethanol was injected via laser technology with the insertion of a
into HCCs under MRI guidance, it could be dis- light-conducting quartz-fiber into a tumor in
158 K.J. Mortele et al

A B

c D

E F
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 159

1983. In 1989 Steger et al (57) described tomatic uterine fibroids in 30 outpatients. The
interstitial laser photocoagulation (ILP) for procedure was well-tolerated by all but one
metastatic liver lesions in two patients. Since patient. Three months after the ablation, the
then, lasers have been used to deliver localized, treated fibroid volume decreased in size by a
controlled heat deposition in multiple organs. mean of 37.5% (range 25% to 49%). At 6
Laser energy can be delivered using different month follow-up, all patients were asympto-
materials such as a neodymium:yttrium- matic and none required further medical or sur-
aluminum-garnet (Nd:YAG), carbon dioxide, gical treatments for the fibroids.
and argon via tiny fiberoptics that are small Laser energy delivered through a stereotac-
enough to be inserted coaxially through an 18- tically guided needle also has been used to
gauge needle (58). Laser light is converted into ablate small breast cancers (62,63). New tech-
heat in the target area, resulting in coagulative nical developments, such as robotic systems
necrosis, secondary degeneration and atrophy, that allow the coordinates of a lesion to be
and tumor shrinkage with minimal damage to approached in a high magnetic field, make a
surrounding structures (58). combination of breast biopsy and subsequent
Early clinical trials using US and CT guid- laser treatment feasible (63).
ance were encouraging, with up to 82% of
treated lesions achieving greater than 50%
Radiofrequency Ablation (RFA)
tumor necrosis (59-61). However, the "zone of
kill" achieved with a single fiberoptic was Percutaneous radiofrequency electrocautery is
limited, requiring multiple fibers and several a thermal ablation technique that causes local
sessions to treat most liver tumors. Diffusing tip tissue destruction by inducing ionic agitation.
fibers, loaded into small catheters, produce This results in heat deposition within the tissue.
larger kill zones. To date, the largest experience Nearly immediate coagulation necrosis is
with MRI-guided laser ablation of tumors has induced at temperatures between 60° and
been reported by Vogi et al (22); using an 100°C. At higher temperatures tissue vaporizes
Nd:YAG laser, 932 liver tumors were treated in and carbonizes. Thus, an essential objective of
335 patients. Computed tomography was used RF ablations is to achieve and maintain tem-
to guide the placement of the catheters. Then, peratures between 50° and 100°C throughout
the patient was moved to a closed MR system the entire target volume for at least 4 to 6
so that the thermal changes during laser depo- minutes (64).
sition could be observed. The local tumor Although this approach was used neurosur-
control rate was 71 % in the first 100 patients, gically for over three decades, Rossi et al (65)
79% in the following 75 patients, and 97% at reported its first use in 13 patients with small
6 months in the remaining patients who were HCCs who were treated with ultrasound-
treated with a cooled power-laser-ablation guided percutaneous RF ablation. Subse-
system. The study demonstrated feasibility and quently, Livraghi et al (66) reported using RFA
utility of monitoring tissue destruction using for metastatic liver tumors. Typically, one or
MRI. more 18- to 21-gauge RF probes are placed
Law and Regan (30) reported their experi- using image-guidance into the lesion. Complete
ence with MRI-guided laser ablation of symp- ablation of lesions measuring 2 to 4cm can be

FIGURE 12.5. MRI-guided cryoablation of rectal mass. (C,D) Intraprocedural axial and sagittal SPGR
cancer recurrence in a patient post-abdominoper- T1-weighted images show cryoprobes within the
ineal resection. (A) Axial FSE T2-weighted MR lesion and enlargement of the iceball. (E,F):
image shows heterogeneous hyperintense mass Gadolinium-enhanced axial and sagittal SPGR Tl-
(arrows) in the presacral area. (B) Gadolinium- weighted MR images obtained 3 months after the
enhanced sagittal SPGR Tl-weighted MR image cryotherapy show subtotal necrosis of the recurrent
shows predominant peripheral enhancement of the rectal cancer.
160 K.J. Mortele et al

achieved (67). New RF probes designs are now ture is elevated due to energy absorption from
available, which include multipronged arrays, the sonic waves, resulting in different degrees
internally cooled electrodes, clustered elec- of thermal damage. Second, there is the phe-
trodes, and saline-cooled electrodes. These nomenon of transient or inertial cavitation.
advances increase the lesion diameter that can Focused ultrasound surgery has two major
be treated (64). advantages: it is less invasive than percutaneous
Because of the interference of RF ablation treatment, as it requires no skin puncture; and
with MRI, multiple strategies have been the ablative effect is not limited by the geome-
attempted to allow MRI to monitor RF abla- try of a probe. Focused ultrasound surgery
tions in real time. Lewin et al (67) reported on ablates a small focal spot-like zone of necrosis.
the first human clinical series of MRI-guided Using a series of FUS ablations, an ablation
RF ablation of abdominal masses. Although RF zone can be achieved that is of virtually any
needle electrodes were placed under MR guid- desired shape. Focused ultrasound surgery can
ance, when MRI was performed the generator be targeted to more complicated lesion geome-
was turned off during imaging, then ablated tries by arranging the multiple sonications to
regions were re-imaged after each session. conform to the target shape. Recently, Wu et al
Using an open configuration MRI system (71) reported their experience in the investiga-
(Magnetom Open; Siemens Medical Solutions; tion of the pathologic changes of HCCs after
Erlangen, Germany), a 50- to 200-W RF gener- FUS treatment. They examined the surgical
ator (Radionics, Inc.; Burlington, MA), and pro- specimens of six patients previously treated
totype MR-compatible 17- x 2-mm shielded with FUS 5 to 18 days earlier, and confirmed
electrodes with a 1- to 3-cm exposed tip, 11 that complete necrosis of the tumors was
tumors were treated in seven patients for a total achieved.
of 13 ablations. There was no significant mor- Focused ultrasound surgery is limited in that
bidity. Four patients showed stable or decreas- it can be applied only in areas where there is no
ing tumor on average follow-up of 152 days. intervening bone or air between the ultrasound
Short-tau inversion recovery (STIR) 1'2- probe and target lesion. However, techniques
weighted sequences were used in between abla- are now being developed to traverse bone (72).
tions for monitoring with RF. This study showed Also, current FUS systems are designed for
that MRI-guided RF ablation of abdominal fixed targets, and therefore applications in the
masses was feasible. Intraprocedural monitor- upper abdomen in which the targets move with
ing with MRI, in which imaging occurs during respiration are difficult. However, just as US is
the treatment, may be possible with a recently used today to image the liver and kidney, FUS
developed switching mechanism that briefly may be used to treat liver and kidney tumors in
interrupts RF deposition during the sampling the future.
for imaging pulse sequences. Ablation duration Uterine fibroids have been one of the first
of a particular lesion is then determined by major areas of clinical application of FUS using
MRI. MR guidance (73). Preliminary experience with
MRI-guided ultrasound surgery has shown that
MRI-guided FUS is feasible for local treatment
Focused Ultrasound Surgery of leiomyomas (Fig. 12.6) (73). The advantage
Focused ultrasound surgery (FUS), or high- of using MRI to guide this form of therapy is
intensity focused ultrasound, is the therapeutic that not only is it excellent for depicting
heat destruction of tissues using US energy that fibroids, but also it allows direct, continuous
is focused on a target deep in the body (68-70). real-time monitoring of the effect. It ensures
Ultrasound beams can be focused and con- that a therapeutic thermal dose has been deliv-
trolled for energy delivery deep into the body ered to the target. If, for example, those changes
without affecting superficial structures. The US are not achieved, repeated sonications in the
beam interacts with tissue at the target volume same area will be possible. Other areas in which
through two mechanisms. First, the tempera- FUS is currently under investigation are the
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 161

FIGURE 12.6. MRI-guided focused ultrasound. (A)


Coronal TSE T2-weighted MR image shows the presence
of a heterogeneous hypointense mass (arrows) consistent
with a fibroid. (B) Intraprocedural axial SPGR image
shows the external ultrasound generator (arrows) with the
patient lying on top of it. Note the absence of any inter-
fering vital structures between the fibroid and the exter-
nal ultrasound generator. (C) Coronal FSE T2-weighted
MR image obtained 3 months after the procedure shows
hyperintense appearance of the ablated area.

B c

treatment of breast fibroadenomas and breast radical prostatectomy, cryotherapy, external


cancer (72). beam radiation therapy, and interstitial radia-
tion therapy (brachytherapy). The latter
involves the placement of radiation seeds
Brachytherapy transperineally. When performed using US
Prostate cancer is the second leading cause of guidance, it is a desirable treatment because it
cancer death in American men (74). Treatment is an outpatient procedure with low morbidity
options for localized prostate cancer include and low cost relative to other treatment options
162 K.J. Mortele et al

(75). However, ultrasound is limited in its prostate brachytherapy improves survival and
ability to define the margins of the prostate cancer control measures (e.g., PSA measure-
gland, urethra, and rectum and in its guidance ments) remains to be established.
of the intended radiation dose to selected por-
tions of the gland.
Magnetic resonance imaging defines the
Microwave Thermocoagulation
prostate gland anatomy and the extent of Microwave (thermo)coagulation is another
prostate cancer better than ultrasound (76). minimally invasive therapy that has been devel-
Therefore, it can be used to improve both radi- oped for the local ablation of tumors. It also
ation dosimetry planning and radiation seed destroys tumor by creating a hyperthermic
placement (Fig. 12.7). With the patient under injury. Several studies have shown the feasibil-
general anesthesia, in the lithotomy position ity of MR-guided microwave therapy in the
and the bladder catheterized, MRI of the treatment of liver tumors (79,80). Morikawa et
prostate gland is first performed using a surface al (79) reported their results in 30 patients with
coil. Then, the prostate, urethra, and rectum liver tumors. All procedures could be success-
are manually contoured on axial TI-weighted fully carried out without complications, and the
images. Using special computer software, therapeutic effects were deemed satisfactory.
prostate gland volumes and the desired radia- Using a notched filter, MRI could be performed
tion dose to each portion of the gland are without electromagnetic interference during
calculated. These dosimetric calculations the ablation.
determine both the number of needles and
number of radiation seeds required to ablate
the peripheral gland that contains the cancer. Conclusions
Then, 1-125 radiation seeds, loaded into 18-
gauge needles, are placed into the prostate Magnetic resonance imaging guidance for
gland at predetermined depths at locations tumor ablation, although still in its early stages
defined by the dosimetric plan. The ability to of development, is generally gaining acceptance
place the seeds precisely is aided both by real- as the field expands to include tumors in deep
time MRI that views the advancing needles and and difficult locations. For example, although
a Plexiglas template that contains rows and a small nonperipheral liver tumor may be fully
columns of holes at 5-mm intervals. This tem- and safely ablated with US or CT guidance,
plate is attached to a rectal obturator, which a large tumor abutting the heart or colon
keeps the template at a fixed position abutting requires meticulous image guidance and moni-
the perineum. It also serves to straighten the toring. Therefore, MRI is needed. Overall, the
anorectal junction, and to delineate the rectal ability of MRI to monitor real-time changes in
wall. Multiplanar MRI is used to confirm accu- tissue temperature suggests MRI as the optimal
rate placement of each needle before each seed method for monitoring tumor ablation using
is deposited. thermal-based energies. The advantages of
The feasibility and safety of MRI-guided diagnostic MRI throughout the body, with its
prostate brachytherapy have been demon- superior ability to detect and characterize
strated previously (77). In this initial report, all lesions and depict anatomy, support its use in
patients were implanted successfully without planning ablations and assessing outcomes.
complication and all patients received 95% or While the feasibility of MRI-guided tumor
more of the ideal radiation dose, a significant ablations has been demonstrated, it is hoped
improvement over the 85 % reported with US- that future clinical trials will demonstrate the
guided methods. In a more recent update (78), added value of MRI and precisely define when
a minimum of 89% coverage of the tumor MRI should be used instead of US or CT.
volume was achieved, while maintaining the Undoubtedly, as the field evolves, more appli-
prostatic urethra and the anterior rectal wall cations will be introduced, and the advantages
below tolerance levels. Whether MRI-guided of MRI should become more apparent.
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 163

A B

c D

FIGURE 12.7. MRI-guided brachytherapy. (A,B) Axial


and sagittal FSE T2-weighted MR images obtained with
the endorectal coil shows diffuse hypointense appear-
ance of the peripheral zone (arrows) of the prostate
gland compatible with prostate carcinoma. (C) Intrapro-
cedural axial SPGR image shows susceptibility artifacts
(arrows) of the radiation seeds deployed within the
prostate gland. (D) Conventional anteroposterior (AP)
radiograph of the pelvis obtained after procedure shows
anatomic distribution of the seeds. (E) Gadolinium-
enhanced axial SPGR T1-weighted MR image obtained
after brachytherapy shows the presence of multiple
seeds (arrows) within the peripheral zone of the
prostate gland. E
164 KJ. Mortele et al

References 14. Daniel BL. Intraprocedural magnetic resonance


imaging-guided interventions in the breast. Top
1. Silverman SG, Jolesz FA, Newman RW, et al. Magn Reson Imaging 2000;11:184-190.
Design and implementation of an interven- 15. Dumoulin CL, Souza P, Darrow RD. Real time
tional MR imaging suite. AJR 1997;168:1465- position monitoring of invasive devices using
1471. magnetic resonance imaging. Magn Reson Med
2. Jolesz FA, Silverman Sa. Interventional mag- 1993;29:411-415.
netic resonance therapy. Semin Intervent Radiol 16. Gohnde SC, Pfammatter T, Steiner P, Erhart P,
1995;12:20-27. Romanowski BJ. MR-guided cholecystectomy:
3. Lu DSK, Lee H, Farahani K, Sinha S, Lufkin R. assessment of biplanar, real-time needle tracking
Biopsy of hepatic dome lesions: semi-real-time in three pigs. Cardiovasc Intervent Radiol 1997;
coronal MR guidance technique. AJR 1997;168: 20:295-299.
737-739. 17. Leung DA, Debatin JF, Wildermuth S, et al.
4. Mueller PR, Stark DD, Simeone JF, et al. MR- Intravascular MR tracking catheter: prelimi-
guided aspiration biopsy: needle design and clin- nary experimental evaluation. AJR 164:1265-
ical trials. Radiology 1986;161:605-609. 1270.
5. Silverman SG, Collick BD, Figueira MR, et al. 18. Wendt M, Busch M, Wetzler R, et al. Shifted
Interactive biopsy in an open configuration MRI rotated keyhole imaging and active tip-tracking
system. Radiology 1995;197:175-181. for interventional procedure guidance. J Magn
6. Frahm YC, Gehl HB, Robberg WA. Technique Reson Imaging 1998;8:251-258.
or MR-guided core biopsy of abdominal masses 19. Glovinski A, Adam G, Buecker A, et al. Catheter
using an open low-field scanner: feasibility and visualization using locally induced actively con-
first clinical results. Fortschr Rontgenstr 1996; trolled field inhomogeneities. Magn Reson Med
164:1:62--67. 1997;38:251-258.
7. Lu DSK, Sinha S, Lucas J, Farahani K, Lufkin R, 20. Glowinski A, Ktirsch J, Adam G, et al. Device
Lewin K MR-guided percutaneus ethanol abla- visualization for interventional MRI using local
tion of liver tissue in a .2-T open MR system: pre- magnetic fields: basic theory and its application
liminary study in porcine model. J Magn Reson to catheter visualization. IEEE Trans Med
Imaging 1997;7:303-308. Imaging 1998;17:786-793.
8. Mahfouz AE, Rahmouni A, Zylbersztejn C, 21. YogI TJ, Mack MG, Muller PK, Straub R,
Mathieu D. MR-guided biopsy using ultrafast Engelmann, Eichler K Interventional MR:
T1- and TI-weighted reordered turbo fast low- interstitial therapy. Eur RadioI1999;9:1479-1487.
angle shot sequences: feasibility and preliminary 22. YogI TJ, Mack MG, Straub R et al. Magnetic
clinical applications. AJR 1996;167:167-169. resonance imaging-guided abdominal interven-
9. Duerk JL, Lewin JS, Wendt M, Petersilge C. tional radiology: laser induced thermotherapy
Remember true-FISP? A high SNR, near 1- of liver metastasis. Endoscopy 1997;29(6):577-
second imaging method for T2-like contrast 583.
interventional MRI at 0.2 T. J Magn Reson 23. Dickinson RJ, Hall AS, Hind AJ, Young IR. Mea-
Imaging 1998;8:203-208. surement of changes in tissue temperature using
10. Papanicolaou N, Eisenberg PJ, Silverman SG, MR imaging. J Comput Assist Tomogr 1986;10:
McNicholas MM, Althausen AF. Prostatic biopsy 468-472.
after proctocolectomy: a transgluteal, CT-guided 24. Kettenbach J, Silverman SG, Hata N, et al.
approach. AJR 1996;166:1332-1334. Monitoring and visualization for MR-guided
11. Kneeshaw PJ, Turnbull LW, Drew PJ. Current laser ablations in an open MR system. J Magn
applications and future direction of MR mam- Reson Imaging 1998;8:933-943.
mography. Br J Cancer 2003;13:4-10. 25. Fosshelm S, Ilyasov K, Wiggen U, et al.
12. Hlawatsch A, Teifke A, Schmidt M, Thelen M. Paramagnetic liposomes as thermosensitive
Preoperative assessment of breast cancer: probes for MRI in vitro feasibility studies. In:
sonography versus MR imaging. AJR 2002;179: Proceedings of the ISMRM Annual Meeting.
1493-1501. Philadelphia: ISMRM, 1999:725.
13. Pflleiderer SO, Reichenbach JR, Azhari, et al. 26. Bartholet A, Goudemant J, Laurent S, et al. Spin
A manipulator system for 14-gauge large core transition molecular materials: intelligent con-
breast biopsies inside a high-field whole-body trast agents for magnetic resonance thermome-
MR scanner. J Magn Reson Imaging 2003;17: try. In: Proceedings of the ISMRM Annual
493-498. Meeting. ISMRM, 2000;138.
12. Magnetic Resonance Imaging Guidance for Tumor Ablation 165

27. Mueller PR, Stark DD, Simeone JF, et al. imaging guided laser induced thermotherapy,
Clinical use of a nonferromagnetic needle for technique and prospective results. Radiology
magnetic resonance-guided biopsy. Gastrointest 1995;196:257-265.
RadioI1989;14:61-64. 42. Livraghi T, Vettori C, Lazzaroni S, et al. Liver
28. Schenck JF, Jolesz FA, Roemer PB, et al. Super- metastases: results of percutaneous ethanol injec-
conducting open configuration MR imaging tion in 14 patients. Radiology 1991;179:709-712.
system for image-guided therapy. Radiology 43. Wong WS, Patel SC, Cruz FS, et al. Cryosurgery
1995;195:805-814. as a treatment for advanced stage hepatocellular
29. Gronemeyer DH, Kaufman L, Rothschild P, et carcinoma. Cancer 1998;82(7):1268-1278.
al. New possibilities and aspects of low-field 44. Brewer WH, Austin RS, Capps GW, et al. Intra-
magnetic resonance tomography. Radiol Diagn operative monitoring and postoperative imaging
1989;30:519-527. of hepatic cryosurgery. Semin Surg Oncol 1998;
30. Law P, Regan L. Interstitial thermo-ablation 14:129-155.
under MRI guidance for the treatment of 45. Weber SM, Lee FT, Warner TF, Chosy SG, Mahvi
fibroids. Curr Opin Obstet Gynecol 2000;12: DM. Hepatic cryoablation: US monitoring of
277-282. extent of necrosis in normal pig liver. Radiology
31. Silverman SG, Tuncali K, Adams DF, et al. MR 1998;207:73-77.
imaging-guided percutaneous cryotherapy of 46. Lee FT, Mahve DM, Chosy SG, et al. Hepatic
liver tumors: initial experience. Radiology 2000; cryosurgery with intraoperative US guidance.
217:657-664. Radiology 1997;202:624---{j32.
32. Lewin JS, Connell CF, Duerek JL, et al. Interac- 47. Steele G Jr. Cryoablation in hepatic surgery.
tive MRI-guided radiofrequency interstitial Semin Liver Dis 1994;14:120-125.
thermal ablation of abdominal tumors: clinical 48. Gilbert JC, Rubinsky B, Roos MS, Wong STS,
trial for evaluation of safety and feasibility. Brennan KM. MR-monitored cryosurgery in
J Magn Reson Imaging 1998;8:40-47. rabbit brain. Magn Reson Imaging 1993;11:
33. Shellock FG, Kanal E. Magnetic Resonance: 1155-1164.
Bioeffects, Safety, and Patient Management. 49. Matsumoto R, Oshio K, Jolesz FA. Monitoring
New York: Raven, 1994:61-69. of laser and freezing-induced ablation in the
34. Hughes CW, Bell C. Anesthesia equipment in liver with Tl-weighted MR imaging. J Magn
remote hospital locations. In: Ehrenwerth J, Reson Imaging 1993;3:770-776.
Eisenkraft JB, eds. Anesthesia Equipment: Prin- 50. Silverman SG, Tuncali K, Adams DF, et al.
ciples and Applications. St. Louis: Mosby, 1993: Percutaneous MR imaging-guided cryotherapy
565-587. of liver metastases. Suppl RadioI1999;213:122.
35. Lewin JS, Duerk JL, Jain JR, et al. Needle local- 51. Shingleton BW, Sewell PE. Percutaneous tumor
ization in MR guided biopsy and aspiration: effect ablation with magnetic resonance imaging guid-
of field strength, sequence design, and magnetic ance. J Urol 2001;165:773-776.
field orientation. AJR 1996;166:1337-1345. 52. vanSonnenberg E, Hadjipavlou A, Chaljub G,
36. Tacke J, Adam G, Speetzen R, et al. MR-guided Nolstie C, Ko E. Therapeutic cryotherapy guided
interstitial cryotherapy of the liver with a novel, by MRI and ultrasound for vascular malforma-
nitrogen-cooled cryoprobe. Magn Reson Med tions in erector muscles of the back. Minim Inva-
1998;39:354-360. sive Ther Allied TechnoI1997;6:343-348.
37. D'Agostino HB, Solinas A. Percutaneous abla- 53. Menendez LR, Tan MS, Kiyabu MT, Chawla SP.
tion therapy for hepatocellular carcinomas. AJR Cryosurgical ablation of soft tissue sarcomas: a
1995;164:1165-1167. phase I trial of feasibility and safety. Cancer
38. Steele G Jr, Ravikumar TS. Resection of hepatic 1999;86:50-57.
metastases from colorectal cancer: biologic per- 54. Goldberg SN, Gazelle GS, Mueller PRo Thermal
spectives. Ann Surg 1989;210:127-138. ablation therapy for focal malignancy: a unified
39. Crews KA, Kuhn JA, McCarty TM, et al. approach to underlying principles, techniques,
Cryosurgical ablation of hepatic tumors. Am J and diagnostic imaging guidance. AJR 2000;174:
Surg 1997;114:614---{j18. 323-331.
40. Gazelle GS, Goldberg SN, Solbiati L, Livraghi T. 55. Shinmoto H, Mulkern RV, Oshio SG, et al. MR
Tumor ablation with radio-frequency energy. appearance and spectral features of injected
Radiology 2000;217:633--646. ethanol in the liver: implication for fast MR-
41. Vogi TJ, Muller PK, Hammerrstingl R, Weinhold guided percutaneous ethanol injection therapy.
N, Felix R. Malignant liver tumors treated with J Comput Assist Torno 1997;21:82-88.
166 K.J. Mortele et al

56. Bown SG. Phototherapy of tumors. World J Surg 69. Yang R, Reilly CR, Rescorla FJ, et al. High-
1983;7:700-709. intensity focused ultrasound in the treatment of
57. Steger AC, Lees WR, Walmsley K, Brown SG. experimental liver cancer. Arch Surg 1991;126:
Interstitial laser hyperthermia: a new approach 1002-1010.
to local destruction of tumours. BMJ 1989;299: 70. Yang R, Sanghvi NT, Rescorla FJ, et al. Extra-
362-365. corporeal liver ablation using sonography-
58. Amin Z, Bown SG, Lees WR. Liver tumor abla- guided high-intensity focused ultrasound. Invest
tion by interstitial laser photocoagulation review RadioI1992;27:796-803.
of experimental and clinical studies. Semin 71. Wu F, Wang Z, Chen W. Pathological study of
Intervent RadioI1993;10:88-100. extracorporeally ablated hepatocellular carci-
59. Amin Z, Bown SG, Lees WR. Local treatment of noma with high-intensity focused ultrasound.
colorectal liver metastases: a comparison of Zhonghua Zhong Liu Za Zhi 2001;23:237-239.
interstitial laser photocoagulation (ILP) and per- 72. Hynynen K, Darkazanli A, Unger E, Schenck JF.
cutaneous alcohol injection (PAl). Clin Radiol MRI-guided noninvasive ultrasound surgery.
1993;48:166-171. Med Phys 1992;20:107-116.
60. Amin Z, Buonaccorsi G, Mills T, et al. Interstitial 73. Tempany CM, Stewart EA, McDermodt N,
laser photocoagulation: evaluation of a 1320nm Quade BJ, Jolesz FA, Hynynen K. MR imaging-
Nd-Yag and an 805nm diode laser: the signifi- guided focused ultrasound surgery of uterine
cance of charring the fibre tip. Lasers Med Sci leiomyomas: a feasibility study. Radiology 2003;
1993;8:113-120. 226:897-905.
61. Amin Z, Donald J, Masters A, et al. Hepatic 74. Wingo PA, Landio S, Reis LAG. An adjustment
metastases: interstitial laser photocoagulation to the 1997 estimate for new prostate cancer
with real-time US monitoring and dynamic CT cases. CA Cancer J Clin 1997;47:239-242.
evaluation treatment. Radiology 1993;187:339- 75. Prestidge BR, Prete JJ, Bucholtz TA, et al. A
347. survey of current clinical practice of permanent
62. Dowlatshahi K, Francescatti DS, Bloom KJ. prostate brachytherapy in the United States. Int
Laser therapy for small breast cancers. Am J J Radiat Oncol BioI Phys 1998;40:461-465.
Surg 2002;184:359-363. 76. Seltzer SE, Getty DJ, Tempany CM, et al. Staging
63. Kaiser WA, Fisher H, Vagner J, Selig M. Robotic prostate cancer with MR imaging: a combined
system for biopsy and treatment of breast lesions radiologist-computer system. Radiology 1997;
in a high-field whole-body magnetic resonance 202:219-226.
tomography unit. Invest Radiol2oo0;35:513-519. 77. D'Amico AV, Cormack R, Tempany CMC, et al.
64. Goldberg SN, Gazelle GS, Dawson S, Rittman W, Real time magnetic resonance image guided
Mueller PR, Rosenthal DI. Tissue ablation with interstitial brachytherapy in the treatment of
radiofrequency: effect of probe size, gauge, dura- selected patients with clinically localized
tion, and temperature on lesion volume. Acad prostate cancer. Int J Radiat Oncol BioI Phys
RadioI1995;2:399-404. 1998;42:507-515.
65. Rossi S, Fornari F, Buscarini L. Percutaneous 78. D'Amico A, Cormack R, Kumar S, Tempany
ultrasound-guided radiofrequency electro- CM. Real-time magnetic resonance imaging-
cautery for the treatment of small hepatocellu- guided brachytherapy in the treatment of
lar carcinoma. J Intervent RadioI1993;8:97-103. selected patients with clinically localized
66. Livraghi T, Goldberg S, Monti F, et al. Saline- prostate cancer. J EndouroI2000;14:367-370.
enhanced radiofrequency tissue ablation in the 79. Morikawa S, Inubushi T, Kurumi, et al. MR-
treatment of liver metastases. Radiology 1997; guided microwave thermocoagulation therapy of
202:205-210. liver tumors: initial clinical experiences using a
67. Lewin JS, Connell CF, Duerek JL, et al. Inter- 0.5 T open MR system. J Magn Reson Imaging
active MRI-guided radiofrequency interstitial 2002;16:576-583.
thermal ablation of abdominal tumors: clinical 80. Naka S, Kurumi Y, Shimizu, et al. Tumor ablation
trial for evaluation of safety and feasibility. with MRI navigation: a novel method of micro-
J Magn Reson Imaging 1998;8:40-47. wave coagulation therapy for hepatic tumour.
68. Hynynen K Damainou CA, Colucci Y, et al. MR Gan To Kagaku Ryoho 2001;28:1591-1594.
monitoring of focused ultrasonic surgery of renal
cortex: experimental and simulation studies.
J Magn Reson Imaging 1995;5:259-266.
13
Magnetic Resonance Imaging
Guidance of Radiofrequency Thermal
Ablation for Cancer Treatment
Daniel T. Boll, Jonathan S. Lewin, Sherif G. Nour, and Elmar M. Merkle

Recent trends in the care of cancer patients soft tissue contrast, vascular conspicuity,
emphasize minimizing invasiveness while thermo- and ligand-sensitive sequences, multi-
improving the effectiveness of treatment by uti- planar imaging capabilities, and lack of radia-
lizing medical resources in a cost-effective tion exposure. These factors make MRI an ideal
manner. This has been evident in the emerging modality for planning, guiding, monitoring, and
and steadily evolving utilization of interven- evaluating cancer treatment procedures. This
tional radiology over the past quarter century. chapter highlights the current utility and future
Minimally invasive interventions performed directions of interventional MRI for use with
by interventional radiologists cover applica- radiofrequency energy in multiple anatomic
tions ranging from straightforward diagnostic regions.
procedures, such as biopsy and aspiration, to a
more complicated array of palliative as well
as therapeutic procedures, such as tumor
embolization, biliary and hollow viscus stent-
General Principles
ing, percutaneous gastrostomy, celiac plexus
nerve blocks, inferior vena cava filter appli-
Radiofrequency Thermal Ablation
cation, and percutaneous image-guided Interstitial RF thermal ablation therapy is an
brachytherapy. While the range of anatomic attractive treatment option for numerous
sites amenable to minimally invasive interven- reasons. This modality has a long and success-
tion steadily increases, oncologic therapy also ful history as it has been utilized in stereotactic
has advanced rapidly; minimally invasive inter- neurosurgical procedures for more than 30
vention, therefore, has assumed a competitive years (10). Neurosurgical indications for this
role beyond the conventional therapeutic form of therapy historically have included cor-
chain of surgery-radiotherapy--ehemotherapy. dotomy, pallidotomy, leukotomy, and thalamot-
Numerous exciting developments in cancer omy for the treatment of intractable pain
treatment have evolved and include gene and involuntary movement disorders (11-14).
therapy (1,2), targeted drug delivery (3,4), Based on data from our site and others, com-
control of angiogenesis (5), and the delivery of plications resulting from RF ablation are
various forms of thermal energy for tumor abla- uncommon as coagulation effects of the heating
tion, such as radiofrequency (RF), laser (6), process contribute to a very low incidence of
focused ultrasound (7), microwave (8), and hemorrhage in the central nervous system
cryotherapy (9). (15,16) and abdomen (17,18).
These developments are expected to profit Reproducible tissue destruction has been ob-
from the substantial contributions of magnetic served in a variety of anatomic structures. Fur-
resonance imaging (MRI), based on its high thermore, both human and animal studies have

167
168 D.T. Boll et al

shown that thermal lesion shape and size can as a treatment and evaluation phase during and
be controlled through electrode design, as well after the ablation.
as the duration and magnitude of the energy The initial planning phase is essential
delivered (19,20). The feasibility of controlling because the ideal electrode trajectory during
energy deposition and allowing gradual tissue the actual procedure is often significantly dif-
heating while performing RF ablations is ferent from that suggested on the preprocedure
demonstrated by a thermistor placed in the imaging data due to the frequent shift of
electrode tip (15). Thermistors continuously anatomic structures when using modified
monitor tissue temperature, while impedance patient positions during the treatment. There-
measurements provide another parameter fore, only preprocedural visualization of the
related to tissue changes at the ablation site. spatial relationships of the anatomic structures,
Unlike radiation therapy but like other localization of the pathology itself, identifica-
thermal ablative therapies, interstitial RF tion of even the smallest blood vessels for vas-
thermal ablation can be repeated numerous cular road-mapping, and the appearance of
times without concern for the cumulative dose. other tumor foci or the accumulation of ascites
The efficacy and safety of RF thermal abla- in the final procedural patient position allow
tion procedures are demonstrated by the wide realistic planning of the optimal ablation elec-
variety of benign and malignant conditions for trode trajectory (Fig. 13.1A,B).
which they can be utilized. The vast majority of The subsequent guidance phase emphasizes
reports describe RF thermal ablation treat- that temporal and spatial resolutions are
ments for liver (21-23) and kidney (24-26) competing objectives in interventional MRI.
tumors, while other target organs in the trunk Continuous visualization, characterized by
include the pancreas (27), prostate (28,29), automated sequential acquisition, reconstruc-
breast (30), spleen (31), and lung (32). tion, and in-room display of multiple image
data sets of highly sensitive anatomic entities,
such as blood vessels and neural structures,
Magnetic Resonance Imaging allows safe and fast interventional device
Guidance of Radiofrequency Thermal advancement and final probe placement, while
Ablation Therapy sparing vital abdominal structures, such as the
Much of the excitement about expanding the gallbladder, bowel loops, and the renal pelvis.
therapeutic uses of RF energy beyond the Once the RF electrode is delivered successfully
neurosurgical, cardiac, and direct intraopera- into the targeted tumor and the interventional-
tive fields, is engendered by the advancements ist is satisfied with the electrode position, the
in imaging technology. The ability to perform deployment of RF energy can be instituted con-
thermal treatment of cancer percutaneously fidently (Fig. 13.1 C).
under direct image guidance has advanced RF The treatment phase represents the greatest
ablation as a minimally invasive option to challenge for any visualization modality. The
surgery that is more suitable for the large sector transformation of unique tissue characteristics
of poor operative candidates. such as density, water and protein content,
The primary principle of image guidance for temperature, as well as fluid vaporization,
ablation electrode-based thermal treatment is microbubble, and gas formation, can be imaged
to secure safe and precise electrode advance- by various devices. However, the correlation
ment into the targeted abnormality, and the of these phenomena with thermal lesion size
subsequent monitoring of the growth and mor- and expected coagulative necrosis is based on
phology of thermally induced coagulative complex estimations.
necrosis from the RF energy. The process of In the early phase of our clinical series, when
image guidance for minimally invasive thera- nonperfused ablation electrodes were used,
peutic procedures such as RF thermal ablation interstitial RF thermal ablation was performed
can be subdivided into a planning and guidance at an electrode tip temperature of 90° ± 2°C.
phase prior to performing the therapy, as well Using this electrode design, lesion length is
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 169

FiGURE 13.1. Hepatic dome metastasis. Transverse as the marked hypointensity developed around the
(A) and coronal (B) turbo spin echo T2-weighted active distal 3 cm of the electrode, surrounded by a
images of the abdomen demonstrate hyperintense rim of hyperintensity reflecting edema and hyper-
metastasis in the posterior dome of the right lobe of emia. Transverse (E) coronal (F) contrast-enhanced
the liver. (C) Turbo spin echo T2-weighted coronal T1-weighted images following interstitial RF thermal
image along the course of the MR-compatible radio- ablation demonstrate hypointensity corresponding
frequency (RF) electrode demonstrates the elec- to avascular area of tumor necrosis. This approxi-
trode positioned within the tumor. (D) Oblique mates the volume of the originally identified tumor,
axial-sagittal turbo spin echo T2-weighted sequence along with a small margin of surrounding normal
noting the effects of interstitial RF thermal ablation parenchyma.

dependent on the exposed conductive tip and carbonization at the electrode-tissue


length, and lesion diameter is limited to approx- interface. This allows energy delivery to regions
imately 2cm (33,34). The limitation to achiev- more distant from the electrode source (35,36).
ing larger lesion diameters is thought to be due Single ablations can be performed with this
to carbonization at the electrode and tissue electrode design that would formerly have
interface, which in turn impairs and finally leads required multiple ablations with intervening
to the cessation of energy transfer. electrode repositioning using a standard RF
With the water-cooled electrode concept, a electrode. To maximize the area of necrosis, we
roller pump is used to circulate chilled water usually use a combination of the cool-tip elec-
inside channels within the electrode to cool the trode with pulsed application of RF energy
tip to 10° to 20°C, thereby preventing charring during which brief periods of current interrup-
170 D.T. Boll et al

tion are triggered automatically when tissue ablation. To date, a continuous effort to further
impedance rises beyond a preset threshold. develop MRI guidance techniques has concen-
Again, the intention is to prevent tissue char- trated mainly on local treatment of cancer and
ring, carbonization, and cavitation, all of which cancer metastases using percutaneous MRI-
lead to cessation of RF current deposition. The guided thermal tumor ablation procedures
ablation time usually ranges from 6 to 20 (Fig. l3.lE,F).
minutes at each electrode location prior to elec-
trode repositioning for total ablation of larger
Interventional MRI Suite
tumors (37,38).
At the conclusion of ablation sessions using The early years of MRI were characterized by
cool-tip electrode designs, a second application relatively long imaging times in closed-bore
of RF energy at the same electrode position may superconducting cylindrical systems. Those
be necessary without cooling, once the desired initial configurations made MRI an unlikely
margins are achieved, to destroy the area adja- guidance and monitoring modality for radio-
cent to the cooled electrode. A practical method logic procedures. Increasingly, many of these
to test the necessity for such additional RF disadvantages have been overcome through
applications is to continue measuring the RF system hardware and pulse sequence improve-
electrode tip temperature for 2 minutes after the ments that have allowed the development of
RF power has been turned off. We reablate the rapid imaging techniques. Therefore, early
center of the annulus-shaped thermal lesion if experiences with thermal ablation procedures
its temperature falls below 60°C before the 2 during which access to the patient was not nec-
minutes have elapsed (Fig. 13.lD). essarily required for the monitoring process,
After the ablation, during the evaluation were gathered in various institutions employ-
phase, high-resolution postprocedure scanning ing conventional cylindrical superconducting
is performed for the final evaluation of thermal systems (40-42).
lesion size and morphology and to exclude pos- The use of MRI for more complex inter-
sible complications in neighboring anatomic ventional procedure guidance includes the
regions. Subsequently, patients at our institu- manipulation of thermal ablation electrodes by
tion are observed overnight before being dis- radiologists. This form of more active interven-
charged the following morning. According to tion requires a departure from conventional
this thermal ablation protocol, we bring the diagnostic concepts and traditional imaging
patient back for follow-up imaging at 2 weeks, systems. The development of an open magnet
and then again at 3 months after ablation, and configuration has facilitated the patient access
at 3-month intervals thereafter. necessary to perform more extensive manipu-
Currently, various imaging modalities are lative maneuvers during interventional proce-
being used for guidance of minimally invasive dures (43-46). Many open MRI system designs
or surgical procedures. However, only MRI have been used to guide percutaneous proce-
combines characteristics such as high soft tissue dures; each system has emphasized the constant
contrast as well as spatial resolution, precise trade-off between field strength and its homo-
vascular conspicuity, multiplanar imaging capa- geneity, as well as the stability of the static and
bilities, and lack of radiation exposure. Fur- gradient magnetic fields that result in direct
thermore, visualization by means of MRI offers effects on the signal-to-noise ratio, access to the
options for temperature mapping, and there- patient, usable field of view, and expense (47).
fore MRI is able to define treatment end points Modern MRI magnets now provide sufficient
by providing immediate feedback regarding the image quality for the interventional guidance
extent of coagulative necrosis during the abla- phase of minimally invasive procedures. Image
tion (39) (Fig. 13.2). With those attributes, MRI acquisition times of 1 to 3 seconds or less
has won increasing acceptance as an interven- per image provide the necessary temporal
tional procedure guidance for aspirations, biop- resolution (48,49). Furthermore, through the
sies, and drainages as well as for monitoring development of in-room, high-resolution,
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 171

FIGURE 13.2. Magnetic resonance thermometry based visualized on T2-weighted magnitude MRI (B), and
on changing Tl relaxation times, diffusion weighting, the tissue damage predicted by direct temperature
and chemical shift phenomenon allows precise measurement, using the chemical shift phenomenon
monitoring of thermal lesion growth, as shown in as imaged on MR phase images (C) in a canine liver
image series (A). Furthermore, strong direct corre- model.
lation is observed between morphologic changes as

RF-shielded monitors, the interventional radi- magnetic field and that create little or no ex-
ologist has the ability to operate the scanner ternal field distortions or image degradation
and view images at the scanner side within the (50,51). Their appearance on the image is due
magnetic fringe field throughout the entire pro- mainly to the displacement of proton-contain-
cedure. In combination with direct patient ing tissue that produces the actual MR signal
access allowed by open imaging systems, this and to the metal intrinsic to the instrument or
capability permits the entire procedure to be paramagnetic markers that is incorporated into
performed with the operator sitting next to the the instrument. These factors lead to the for-
patient throughout the procedure (Fig. 13.3). mation of areas of increased susceptibility, and
In addition, thermal ablation electrodes have therefore contribute to areas of signal void in
been developed that are undeflected by the the images (52).
172 D.T. Boll et al

Safety Issues for Interventional


MRI During Radiofrequency
Energy Deposition
In contrast to widespread imaging modalities
used for interventional guidance, such as ultra-
sound and computed tomography (CT), which
interact with human tissue and the interven-
tional device only during the actual visualiza-
tion process, a major basic difference of
MRI is its continuous strong magnetic field.
Although risks are less prominent with the low
and medium magnetic field strengths of 0.2 to
0.5T typically used for MRI-guided proce-
dures, hazardous consequences can result when
ferromagnetic instruments become attracted to
and are accelerated in the fringe field of the
MRI unit. Serious or even fatal injuries can
occur. Therefore, the generally accepted rule
FIGURE 13.3. C-arm system for percutaneous inter- is that no ferromagnetic materials should be
vention. For immediate monitoring of thermal brought within the 5-gauss line of any MR
ablation electrode placement and subsequent RF scanner. Electric burns can result from direct
thermal ablation acquired breath-hold images are
electromagnetic induction in a conductive loop,
presented to the interventionalist on a shielded
liquid crystal display (LCD) monitor (arrow) adja-
induction in a resonant conducting loop, or
cent to the scanner. A computer mouse on the LCD electric field resonant coupled with a wire
console and foot pedals (not shown) allow the (55,56). Acoustic noise during interventions on
scanner to be operated by the radiologist throughout open low- and medium-field scanners normally
the procedure. does not reach the occupational exposure limit
of 15 minutes per day at 115 dB.
Adequate visualization of the target pathol-
ogy and surrounding anatomy is crucial, espe-
The modification and development of rapid cially in interventional MRI. Because of the
gradient-echo pulse sequences that facilitate a need for high temporal resolution during
wide range of tissue contrast in a time frame the advancement and final placement of the
sufficient for device tracking even at low field thermal ablation electrode, the resultant images
strength and with the suboptimal coil position do not have the quality expected from a purely
sometimes required to access a puncture site, diagnostic sequence. Different, nearly real-time
have proven their capability and usefulness in pulse sequences are available, thus allow-
interactive guidance procedures during device ing multiple tissue contrasts to be obtained,
placement. Furthermore, the inherent inability depending on the sequence design as well as the
of MRI to monitor actively a thermal lesion as implemented parameters (57).
it is created, due to imaging interference caused The most commonly used sequences for the
by the RF source, has been overcome. New guidance phase for MRI of RF thermal abla-
software and hardware modifications have tions are fast imaging with steady-state pre-
been developed that allow RF energy to be cession (FISP) techniques, with a temporal
deposited during imaging with short interrup- resolution of 1 to 3 seconds per image, FISP
tions during the sampling periods, thereby sequences with balanced gradients in all spatial
maintaining tissue temperature, while making directions (true FISP), characterized by an
interference-free real-time monitoring possible acquisition speed of 1 second per image, and
(53,54). finally a time-reversed version of the FISP se-
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 173

quence (known as PSIF) with a temporal reso- Economic Considerations


lution of 4 to 5 seconds per image. However,
those gradient echo sequences are associated The cost-effectiveness of interventional MRI
with more prominent susceptibility artifacts has been debated primarily because conven-
from thermal ablation electrodes than with the tional, less expensive, and widely available
relatively slower spin echo or turbo spin echo imaging modalities seemed adequate in the
sequences. Therefore, to reduce artifactual past. Compared to current ultrasound and
needle widening, the use of turbo spin echo CT-guided procedures, MRI guidance is still
imaging for position confirmation should be slightly more expensive. However, when
strongly considered when electrode placement thermal therapy guidance, monitoring, and
within 5mm of a major neurovascular structure evaluation are the issues, the unparalleled
is contemplated (58). ability of MRI to detect residual untreated
After successful placement of the thermal tumor foci during the ablation session out-
ablation electrode, MRI provides incomparable weighs the extra cost associated with the use of
techniques for monitoring lesion formation and this technology. In fact, in many institutions in
morphology. The development of temperature- which thermal therapy is performed under
sensitive MR sequences enables accurate ultrasound or CT guidance, the patient is ulti-
online monitoring of heat deposition (59). The mately transferred to the MR scanner follow-
relationship of MR signal intensity change to ing ablation to confirm the extent of necrosis.
tissue temperature is a complex phenomenon, Compared to the surgical approach, the use
and precise MR temperature measurement is of MRI guidance for RF thermal ablations is
difficult. However, the phase transition from associated with tremendous cost reduction,
viable to necrotic tissue can be imaged directly from a decrease in the cost of the procedure
using changes in the tissue relaxation parame- itself relative to surgery, to the equal or greater
ters, Tl and T2, that occur in the process of savings realized by avoiding the intensive care
necrosis formation (60,61). unit as well as routine hospitalization. This is in
When applying the relatively high currents addition to the patient benefits that result from
necessary to perform RF ablation procedures the reduction in morbidity, mortality, post-
successfully, system grounding through an procedure discomfort, and recovery time when
adequate surface area is crucial. This is due to open surgical procedures can be avoided (63).
the fact that when RF current passes through a
patient's body and closes its complete electric
circuit, an equal amount of current is deposited
Applications
at the return electrodes, such as the grounding
pad, compared to the source electrode (62). The
MRI-Guided Radiofrequency Thermal
optimal outcome of an RF ablation session is
Ablation of the Liver
the generation of a thermal lesion that covers Over the past decade, hepatic malignancies,
the whole targeted tumor plus a safety margin such as primary hepatocellular carcinomas (17)
comparable to the 0.5- to I-em rim generally and hepatic metastasis (64), have evolved as
targeted during surgery. While undertreatment prime abdominal therapeutic sites amenable to
is obviously an unacceptable outcome, over- treatment with RF-induced thermal ablation.
ablation also is not free from risks. Injury of vital The anatomic structure of the liver capsule and
structures adjacent to the target tumor can com- the liver parenchyma accounts for the relatively
plicate the treatment. In addition to the concern low incidence of complications, such as bleed-
about collateral damage, it is also important ing following insertion of interventional devices
when planning extensive ablations to consider of various calibers, and therefore substantiate
whether sufficient organ function can be pre- the rationale to use hepatic RF thermal abla-
served and to be aware that large-volume tissue tion in the liver (65).
necrosis is associated with a higher incidence of Prior to the clinical application of this tech-
infection and the postablation syndrome. nique, however, a representative small animal
174 nT. Boll et al

model focusing on implantation, harvesting, Experience gathered during the develop-


and subsequent MRI-guided RF thermal abla- ment phase of the hepatic RF thermal ablation
tion treatment in the rabbit liver was developed technique in the animal model is of paramount
at our institution (21) (Fig. 13.4). It demon- importance for establishing this form of treat-
strated that the absence of major interventional ment within the clinical routine environment
complications, the mean treatment duration of (Fig. 13.5). The efficacy and success of percuta-
44 minutes including all evaluation by MRI, neous RF thermal ablation can be diminished
and the ability to monitor tissue damage during by a tumor mass of more than 30% of the com-
and immediately after the procedure confirmed plete liver volume, extrahepatic tumor exten-
the feasibility of this approach. Important infor- sion, or severe dilatation of the intrahepatic
mation concerning the correlation of image biliary system.
appearance of the induced coagulative necrosis The probability of a successful outcome
and gross pathologic evaluation revealed a dif- with MRI-guided RF ablation increases with
ference of less than 2 mm. T2-weighted imaging smaller tumor size, less than 3 to 4 cm. Studies
continuously overestimated the lesion size, performed in our institution showed that TI-
consistent with other studies (66). Remaining weighted turbo spin echo and short-tau inver-
tumor within the liver was best identified on TI- sion recovery (STIR) sequences utilized after
weighted turbo spin echo sequences; these the RF thermal ablation demonstrate the
results followed those of animal studies per- zone of tissue damage with similar precise
formed at other anatomic locations (67,68). conspicuity compared to contrast-enhanced
Follow-up examinations consisting of T2- T1-weighted spin echo sequences. Especially
weighted turbo spin echo sequences and intra- the contrasts within the thermally induced
venous gadopentetate dimeglumine-enhanced lesion, the edematous surrounding hyperin-
T1-weighted imaging were able to demonstrate tense rim, and the physiologic surrounding liver
an accuracy of estimating RF thermal lesion tissue imaged with T2-weighted turbo spin echo
size within 2 to 3 mm. and STIR sequences are comparable to con-

A I
FIGURE 13.4. Small animal model demonstrating the
feasibility of MRI-guided liver ablation in a rabbit
B
measurements on gross pathology (B) and histology
(see ref. 66). Animal experiments in multiple organ
specimen. Area of low signal intensity (A) (arrow- systems were used to document the ability of this
heads) on T2-weighted image developing around method of imaging tissue damage to be used to tailor
electrode during RF ablation correlates closely with the treatment phase ofMRI-guided thermal ablation.
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 175

FIGURE 13.5. Images of a 74-year-old patient with contrast-enhanced images obtained after ablation
three known metastases from a previously resected and removal of the electrode demonstrate a central
leiomyosarcoma of the stomach. T2-weighted axial low- signal necrotic core replacing the bright tumor
image (A) through the right lobe of the liver demon- nodule, with a thin, bright rim of surrounding edema.
strates a I-em bright tumor nodule (arrow). T2- By tailoring this region of tumor necrosis as depicted
weighted coronal image (B) demonstrates the on MRI, lesions of variable size and shape can be
titanium electrode identified as a dark band with its effectively ablated along with a margin of surround-
tip through the center of the tumor nodule (arrow). ing tissue.
Transverse (C) and coronal (D) Tl-weighted

trast media-enhanced sequences. Thus repeat benefited from the earlier detection of smaller
images are obtained during the thermal RF masses (71,72). With the increasing ability to
ablation, because of the contrast media inde- detect renal malignancies, nephron-sparing
pendence of these sequences (69). therapies have been developed that are aimed
at the preservation of renal function. This
preservation is of paramount importance in an
MRI -Guided Radiofrequency Thermal
anatomically or functionally solitary kidney or
Ablation of Kidney Tissue and Tumors in conditions such as Von Rippel-Lindau
The proliferation of cross-sectional imaging disease linked to the occurrence of recurrent
techniques has led to an increase in the inci- renal cell carcinomas. Studies have demon-
dental visualization and therefore early detec- strated that minimally invasive and nephron-
tion of renal malignancies (70). Furthermore, sparing treatments of small renal malignancies
high-risk patients who underwent cross- have the same efficacy as radical nephrectomy
sectional imaging for suspected cancer have (73-75). Minimally invasive, image-guided
176 D.T. Boll et al

FIGURE 13.6. Animal model presenting the feasibil- short-tau inversion recovery (STIR) (B), as well as
ity of MRI-guided kidney ablation and the correla- contrast-enhanced T1-weighted MRI (C).
tion with porcine gross pathology specimen (A) and

renal tissue ablation has been described using 100% of cases during cryoablation (80). The
techniques such as cryotherapy, focused ultra- procedure time required to perform interven-
sound, and microwave application (76-79). tional thermal ablation in the kidney was mea-
Radiofrequency thermal energy to treat sured at a mean of 11 minutes. By employing
malignant lesions within the kidneys was not MRI during the procedure, the growth and
described prior to the development of an morphology of the coagulative necrosis were
animal model to prove the feasibility and safety monitored successfully, allowing repositioning
of this approach (24) (Fig. 13.6). Under MRI of the ablation electrode immediately. It was
guidance, the desired insertion site in the proven that MRI succeeded in predicting the
retroperitoneal kidneys was accessed without size and morphology of an induced thermal
difficulty with the RF ablation probe. The excel- lesion within porcine kidneys.
lent vessel conspicuity of MRI facilitates the At our institution, a phase II clinical trial
avoidance of large vessels during the placement focusing on interactive MRI-guided interstitial
of the ablation electrode. However, due to the RF thermal ablations of primary kidney tumors
high vascularity of the targeted site, the devel- was initiated with the purpose of evaluating
opment of small puncture-related hematomas long-term follow-up examinations of a cohort
was observed in 30% of porcine kidney RF of patients (Fig. 13.7). The trial specifically
ablations. However, small hemorrhages imme- was designed to demonstrate efficacy of this
diately visualized by MRI are not an uncom- treatment modality and to analyze patient
mon complication of interstitial thermotherapy discomfort or any complications associated
in the kidney, with an occurrence rate up to with this procedure. Complete ablation of renal
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 177

masses was achieved in 93 % of all patients Patients with complete coagulative necrosis
treated and in 100% of patients with tumors of of their renal malignancy showed no clinical or
4cm diameter or less, based on postprocedure imaging evidence of tumor recurrence with a
and follow-up examinations on T2-weighted mean follow-up of 366 ± 129 days. On volu-
and STIR images. Due to the proximity of the metric analysis, it was demonstrated that the
developing thermal lesion to the renal pelvis, thermal lesion increased in size by approxi-
further RF application could not be performed mately 2mL on the 2-week follow-up examina-
in one larger tumor in which a small rim of tion, compared to the images obtained at the
hyperintense material was still visible on T2- conclusion of the ablation session. Subse-
weighted and STIR images. In 40% of all quently, the lesion volume slowly decreased
treated patients, oral analgesia was adminis- during the follow-up period. Signal intensity
tered on the evening after the procedure; remained low on T2-weighted and STIR images
however, none of these patients required pain throughout the follow-up period for all suc-
medication on discharge. cessful ablation procedures. The ability to
detect inadequately treated portions of the
renal malignancy and interactively reposition
the electrode during therapy was critical in this
investigation. Despite the relatively small sizes
of the masses, manipulation of the electrode
into untreated tumor was necessary in 80% of
the treated patients to achieve complete tumor
ablation. This relatively high proportion is
undoubtedly related to the high vascularity
of both the tumor and the adjacent renal
parenchyma.
With mean follow-up examinations of
greater than 1 year, this clinical study suggests
that interactive MRI-guided RF renal ablation
for treatment of primary renal tumors has a
high success rate to achieve complete ablation
and a low level of tumor recurrence; it is also
extremely well tolerated by patients (81).

Research and Future Directions


The feasibility and safety of RF thermal abla-
tions performed under MRI guidance for other
body organs have been investigated in animal
models. Multiple reports have proven the ap-
plicability of this technique to more than the
FIGURE 13.7. Follow-up examination 20 months after current clinically recognized applications. Posi-
RF thermal ablation of the right kidney in a patient
tive results from pancreatic (27), long bone
with renal cell carcinoma. T2-weighted (A) and
STIR (B) coronal images demonstrate typical encas-
(82), and vertebral (83) ablation sites (Fig. 13.8)
ing scar of a prior successfully ablated exophytic promise to expand the future of cancer treat-
renal cell carcinoma. On turbo spin echo (TSE) T2- ment by taking advantage of the minimally
weighted images (A), the fat surrounding the shrink- invasive nature of RF ablation along with the
ing low-signal tumor scar remains bright, but is superb value of MRI for guiding and monitor-
suppressed on STIR images (B). ing RF thermal treatment.
178 nT. Boll et al

tracking optimIzation. Interactive MRI scan


plane definition for rigid interventional devices,
such as RF thermal electrodes, without the
need for stereotactic cameras, is now possible
using wireless, tuned fiducial markers mounted
to the device (85).

References
1. Allport JR, Weissleder R. In vivo imaging of
gene and cell therapies. Exp Hematol 2001;
29(11):1237-1246.
2. Yang X,Atalar E, Li D, et al. Magnetic resonance
imaging permits in vivo monitoring of catheter-
based vascular gene delivery. Circulation 2001;
104(14):1588-1590.
3. Guerquin-Kern JL, Volk A, Chenu E, et al.
Direct in vivo observation of 5-fluorouracil
release from a prodrug in human tumors hetero-
transplanted in nude mice: a magnetic resonance
study. NMR Biomed 2000;13(5):306-310.
4. Calvo BF, Semelka RC. Beyond anatomy: MR
imaging as a molecular diagnostic tool. Surg
Oncol Clin North Am 1999;8(1):171-183.
5. Fuss M, Wenz F, Essig M, et al. Tumor angiogen-
esis of low-grade astrocytomas measured by
dynamic susceptibility contrast-enhanced MRI
(DSC-MRI) is predictive of local tumor control
after radiation therapy. Int J Radiat Oncol BioI
FIGURE 13.8. The feasibility and safety of RF Phys 2001;51(2):478-482.
thermal ablation performed under MRI guidance 6. Mack MG, Straub R, Eichler K, et al. [Needle
also have been investigated and successfully per- holder for reducing radiation burden of examin-
formed in animal models in other body organs such ers in CT-assisted puncture. Technical contribu-
as the pancreas (A) and long bone (B). tion]. Radiologe 2001;41(10):927-929.
7. Huber PE, Jenne JW, Rastert R, et al. A new
noninvasive approach in breast cancer ther-
apy using magnetic resonance imaging-guided
Further development of the RF technology focused ultrasound surgery. Cancer Res 2001;
will be necessary to create larger thermal 61(23):8441-8447.
lesions with the least number of RF electrode 8. Chen JC, Moriarty JA, Derbyshire JA, et al.
insertions. This is of paramount importance for Prostate cancer: MR imaging and thermometry
reducing such risks as tumor seeding, bleeding, during microwave thermal ablation-initial ex-
and infection, and is also important to reduce perience. Radiology 2000;214(1):290-297.
the procedure time and thus improve patient 9. Mala T, Samset E, Aurdal L, Gladhaug I, Edwin
compliance, particularly when managing large B, Soreide 0. Magnetic resonance imaging-
estimated three-dimensional temperature distri-
tumors. Techniques already investigated in-
bution in liver cryolesions: a study of cryolesion
clude the use of multiple-probe RF electrode characteristics assumed necessary for tumor
arrays that allow a higher maximum energy to ablation. Cryobiology 2001;43(3):268-275.
be deposited into the tissue (36,84). 10. Brodkey JS, Miyazaki Y, Ervin FR, Mark VB.
Ongoing extensive interventional MR Reversible heat lesions with radiofrequency
physics research is primarily focused on MR current: a method of stereotactic localization.
pulse sequence development (57) and device J Neurosurg 1964;21:49-53.
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 179

11. Tew JM Jr, Keller JT. The treatment of trigemi- the kidney in a porcine model. AJR 1999;173(3):
nal neuralgia by percutaneous radiofrequency 645-651.
technique. Clin Neurosurg 1977;24:557-578. 25. Pavlovich CP, Walther MM, Choyke PL, et al.
12. Hitchcock ER, Teixeira MJ. A comparison of Percutaneous radiofrequency ablation of small
results from center-median and basal thalamot- renal tumors: initial results. J Urol 2002;167(1):
omies for pain. Surg NeuroI1981;15(5):341-351. 10-15.
13. Laitinen LV, B ergenheim AT, Hariz MI. Leksell's 26. Lewin JS, Connell CD, Duerk JL, Nour SG,
posteroventral pallidotomy in the treatment Resnick MI, Haaga JR. A phase II clinical trial
of Parkinson's disease. J Neurosurg 1992;76(1): of interactive MR-guided interstitial radio-
53-61. frequency thermal ablation of primary kidney
14. Rosomoff HL, Brown CJ, Sheptak P. Percuta- tumors: preliminary results. Proceedings of the
neous radiofrequency cervical cordotomy: tech- Radiological Society of North America 87th
nique. J Neurosurg 1965;23(6):639-644. Meeting, 2001:261.
15. Zervas NT, Kuwayama A. Pathological charac- 27. Merkle EM, Haaga JR, Duerk JL, Jacobs GH,
teristics of experimental thermal lesions. Com- Brambs HJ, Lewin JS. MR imaging-guided radio-
parison of induction heating and radiofrequency frequency thermal ablation in the pancreas in a
electrocoagulation. J Neurosurg 1972;37(4):418- porcine model with a modified clinical C-arm
422. system. Radiology 1999;213(2):461-467.
16. Farahani K, Mischel PS, Black KL, De Salles 28. Syed AH, Stewart LH, Hargreave TB. Day-case
AA, Anzai Y, Lufkin RB. Hyperacute thermal local anaesthetic radiofrequency thermal abla-
lesions: MR imaging evaluation of development tion of benign prostatic hyperplasia: a four-year
in the brain. Radiology 1995;196(2):517-520. follow-up. Scand J Urol Nephrol 2000;34(5):
17. Rossi S, Di Stasi M, Buscarini E, et al. Percuta- 309-312.
neous RF interstitial thermal ablation in the 29. Zlotta AR, Djavan B, Matos C, et al. Percuta-
treatment of hepatic cancer. AJR 1996;167(3): neous transperineal radiofrequency ablation of
759-768. prostate tumour: safety, feasibility and patholog-
18. Lewin JS, Connell CF, Duerk JL, et al. Inter- ical effects on human prostate cancer. Br J Urol
active MRI-guided radiofrequency interstitial 1998;81(2):265-275.
thermal ablation of abdominal tumors: clinical 30. Izzo F, Thomas R, Delrio P, et al. Radiofrequency
trial for evaluation of safety and feasibility. ablation in patients with primary breast car-
J Magn Reson Imaging 1998;8(1):40-47. cinoma: a pilot study in 26 patients. Cancer
19. Zervas NT. Eccentric radio-frequency lesions. 2001 ;92(8):2036-2044.
Confin NeuroI1965;26(3):143-145. 31. Wood BJ, Bates S. Radiofrequency thermal
20. Chung YC, Duerk JL, Lewin JS. Generation and ablation of a splenic metastasis. J Vase Intervent
observation of radiofrequency thermal lesion RadioI2001;12(2):261-263.
ablation for interventional magnetic resonance 32. Dupuy DE, Zagoria RJ,Akerley W, Mayo-Smith
imaging. Invest Radiol 1997;32(8):466-474. WW, Kavanagh PV, Safran H. Percutaneous
21. Merkle EM, BoIl DT, Boaz T, et al. MRI-guided radiofrequency ablation of malignancies in the
radiofrequency thermal ablation of implanted lung. AJR 2000;174(1 ):57-59.
VX2 liver tumors in a rabbit model: demonstra- 33. McGahan Jp, Gu WZ, Brock JM, Tesluk H, Jones
tion of feasibility at 0.2 T. Magn Reson Med CD. Hepatic ablation using bipolar radiofre-
1999;42(1 ):141-149. quency electrocautery. Acad Radiol 1996;3(5):
22. Nicoli N, Casaril A, Marchiori L, Mangiante G, 418-422.
Hasheminia AR. Treatment of recurrent hepato- 34. Rossi S, Buscarini E, Garbagnati F, et al. Percu-
cellular carcinoma by radiofrequency thermal taneous treatment of small hepatic tumors by
ablation. J Hepatobiliary Pancreat Surg 2001; an expandable RF needle electrode. AJR 1998;
8(5):417-421. 170(4):1015-1022.
23. Buscarini L, Buscarini E, Di Stasi M, Vallisa D, 35. Goldberg SN, Gazelle GS, Solbiati L, Rittman
Quaretti P, Rocca A. Percutaneous radiofre- WJ, MueIler PRo Radiofrequency tissue ablation:
quency ablation of small hepatocellular carci- increased lesion diameter with a perfusion elec-
noma: long-term results. Eur Radiol 2001;11(6): trode. Acad Radiol 1996;3(8):636-644.
914-921. 36. Lorentzen T. A cooled needle electrode for
24. Merkle EM, Shonk JR, Duerk JL, Jacobs GH, radiofrequency tissue ablation: thermodynamic
Lewin JS. MR-guided RF thermal ablation of aspects of improved performance compared
180 D.T. Boll et al

with conventional needle design. Acad Radiol 50. Ladd ME, Erhart P, Debatin JF, Romanowski BJ,
1996;3(7):556-563. Boesiger P, McKinnon Gc. Biopsy needle sus-
37. Solbiati L, Goldberg SN, Ierace T, et al. Hepatic ceptibility artifacts. Magn Reson Med 1996;
metastases: percutaneous radio-frequency abla- 36(4):646-651.
tion with cooled-tip electrodes. Radiology 1997; 51. Lewin JS, Duerk JL, Jain VR, Petersilge CA,
205(2):367-373. Chao CP, Haaga JR. Needle localization in MR-
38. Solbiati L, Ierace T, Goldberg SN, et ai. Percuta- guided biopsy and aspiration: effects of field
neous US-guided radio-frequency tissue abla- strength, sequence design, and magnetic field
tion of liver metastases: treatment and follow-up orientation. AJR 1996;166(6):1337-1345.
in 16 patients. Radiology 1997;202(1):195-203. 52. Bakker CJ, Smits HF, Bos C, et al. MR-guided
39. Chung YC, Duerk JL, Shankaranarayanan A, balloon angioplasty: in vitro demonstration of
Hampke M, Merkle EM, Lewin JS. Temperature the potential of MRI for guiding, monitoring, and
measurement using echo-shifted FLASH at evaluating endovascular interventions. J Magn
low field for interventional MRI. J Magn Reson Reson Imaging 1998;8(1):245-250.
Imaging 1999;9:138-145. 53. Zhang Q, Chung YC, Lewin JS, Duerk JL. A
40. Kahn T, Harth T, Bettag M, et al. Preliminary method for simultaneous RF ablation and MRI.
experience with the application of gadolinium- J Magn Reson Imaging 1998;8(1):110-114.
DTPA before MR imaging-guided laser-induced 54. Patel KC, Duerk JL, Zhang Q, et al. Methods for
interstitial thermotherapy of brain tumors. providing probe position and temperature infor-
J Magn Reson Imaging 1997;7(1):226-229. mation on MR images during interventional
41. Vogi TJ, Mack MG, Straub R, Roggan A, Felix R. procedures. IEEE Trans Med Imaging 1998;
Magnetic resonance imaging-guided abdominal 17(5):794-802.
interventional radiology: laser-induced ther- 55. Dempsey MF, Condon B, Hadley DM. Investi-
motherapy of liver metastases. Endoscopy 1997; gation of the factors responsible for burns during
29(6):577-583. MRI. J Magn Reson Imaging 2001;13(4):627-
42. Liu H, Martin AJ, Truwit CL. Interventional MRI 631.
at high-field (1.5 T): needle artifacts. J Magn 56. Nitz WR, Oppelt A, Renz W, Manke C, Lenhart
Reson Imaging 1998;8(1):214-219. M, Link 1. On the heating of linear conductive
43. Lewin JS, Nour SG, Duerk JL. Magnetic reso- structures as guide wires and catheters in inter-
nance image-guided biopsy and aspiration. Top ventional MRI. J Magn Reson Imaging 2001;
Magn Reson Imaging 2000;11(3):173-183. 13(1):105-114.
44. Lufkin R. Teresi L, Hanafee W. New needle for 57. Duerk JL, Butts K, Hwang Kp, Lewin JS. Pulse
MR-guided aspiration cytology of the head and sequences for interventional magnetic resonance
neck. AJR 1987;149(2):380-382. imaging. Top Magn Reson Imaging 2000;11(3):
45. Gronemeyer DH, Kaufman L, Rothschild P, 147-162.
Seibel RM. [New possibilities and aspects of low- 58. Lewin JS, Petersilge CA, Hatem SF, et al. Inter-
field magnetic resonance tomography]. Radiol active MR imaging-guided biopsy and aspiration
Diagn (Berl) 1989;30(4):519-527. with a modified clinical C-arm system. AJR 1998;
46. Lewin JS. Interventional MR imaging: concepts, 170(6):1593-1601.
systems, and applications in neuroradiology. 59. Botnar RM, Steiner P, Dubno B, Erhart P, von
AJNR 1999;20(5):735-748. Schulthess GK, Debatin JE Temperature quan-
47. Hinks RS, Bronskill MJ, Kucharczyk W, tification using the proton frequency shift tech-
Bernstein M, Collick BD, Henkelman RM. nique: in vitro and in vivo validation in an open
MR systems for image-guided therapy. J Magn 0.5 tesla interventional MR scanner during RF
Reson Imaging 1998;8(1):19-25. ablation. J Magn Reson Imaging 2001;13(3):
48. Duerk JL, Lewin JS, Wendt M, Petersilge C. 437-444.
Remember true FISP? A high SNR, near 60. Matsumoto R, Oshio K, Jolesz FA. Monitoring
I-second imaging method for T2-like contrast of laser and freezing-induced ablation in the
in interventional MRI at .2 T. J Magn Reson liver with Tl-weighted MR imaging. J Magn
Imaging 1998;8(1):203-208. Reson Imaging 1992;2(5):555-562.
49. Chung YC, Merkle EM, Lewin JS, Shonk JR, 61. Bleier AR, Jolesz FA, Cohen MS, et al. Real-time
Duerk JL. Fast T(2)-weighted imaging by PSIF magnetic resonance imaging of laser heat depo-
at 0.2T for interventional MRI. Magn Reson sition in tissue. Magn Reson Med 1991;21(1):
Med 1999;42(2):335-344. 132-137.
13. Magnetic Resonance Imaging Guidance of Radiofrequency Thermal Ablation 181

62. Goldberg SN, Solbiati L, Halpern EF, Gazelle 75. Licht MR, Novick AC, Goormastic M. Nephron
GS. Variables affecting proper system grounding sparing surgery in incidental versus suspected
for radiofrequency ablation in an animal model. renal cell carcinoma. J UrolI994;152(1):39-42.
J Vase Intervent RadioI2000;11(8):1069-1075. 76. Kigure T, Harada T, Yuri Y, Satoh Y, Yoshida K.
63. Lewin JS. MR guides intervention and keeps Laparoscopic microwave thermotherapy on
costs down. Diagn Imaging (San Franc) 1998; small renal tumors: experimental studies using
Suppl Open MRI:MR22-MR24. implanted VX-2 tumors in rabbits. Eur Urol
64. Livraghi T, Goldberg SN, Monti F, et al. Saline- 1996;30(3):377-382.
enhanced radio-frequency tissue ablation in the 77. Lotfi MA, McCue P, Gomella LG. Laparoscopic
treatment of liver metastases. Radiology 1997; interstitial contact laser ablation of renal lesions:
202(1 ):205-210. an experimental model. J Endourol 1994;8(2):
65. Wildhirt E, Moller E. [Experience with nearly 153-156.
20,000 blind liver punctures]. Med Klin 1981; 78. Watkin NA, Morris SB, Rivens IH, ter Haar GR.
76(9):254-255,279. High-intensity focused ultrasound ablation of
66. Boaz TL, Lewin JS, Chung YC, Duerk JL, the kidney in a large animal model. J Endourol
Clampitt ME, Haaga JR. MR monitoring of 1997;11(3):191-196.
MR-guided radiofrequency thermal ablation of 79. Uchida M, Imaide Y, Sugimoto K, Uehara H,
normal liver in an animal model. J Magn Reson Watanabe H. Percutaneous cryosurgery for renal
Imaging 1998;8(1):64-69. tumours. Br J UroI1995;75(2):132-136.
67. Anzai Y, Lufkin R, DeSalles A, Hamilton DR, 80. Chosy SG, Nakada SY, Lee FT Jr, Warner TF.
Farahani K, Black KL. Preliminary experience Monitoring renal cryosurgery: predictors of
with MR-guided thermal ablation of brain tissue necrosis in swine. J Urol 1998;159(4):
tumors. AJNR 1995;16(1):39-48. 1370-1374.
68. Anzai Y, Brunberg JA, Lufkin RB. Imaging of 81. Lewin J, Nour S, Connell C, Sulman A, Duerk J,
nodal metastases in the head and neck. J Magn Resnick MI. Followup Findings of a Phase II
Reson Imaging 1997;7(5):774-783. Clinical Trial of Interactive MR-Guided Inter-
69. Aschoff AJ, Rafie N, Jesberger JA, Duerk JL, stitial Radiofrequency Thermal Ablation of
Lewin JS. Thermal lesion conspicuity following Primary Kidney Tumors. Proceedings of the
interstitial radiofrequency thermal tumor abla- International Society of Magnetic Resonance in
tion in humans: a comparison of STIR, turbo Medicine 2002;1:740.
spin-echo T2-weighted, and contrast-enhanced 82. Aschoff AJ. Merkle EM, Emancipator SN,
Tl-weighted MR images at 0.2T. J Magn Reson Petersilge CA, Duerk JL, Lewin JS. Femur:
Imaging 2000;12(4):584-589. MR imaging-guided radio-frequency ablation in
70. Smith SJ, Bosniak MA, Megibow AJ, Hulnick a porcine model-feasibility study. Radiology
DH, Horii SC, Raghavendra BN. Renal cell car- 2002;225(2):471-478.
cinoma: earlier discovery and increased detec- 83. Nour SG, Aschoff AJ, Mitchell IC, Emancipator
tion. Radiology 1989;170(3 pt 1):699-703. SN, Duerk JL, Lewin JS. MR imaging-guided
71. Bosniak MA, Rofsky NM. Problems in the radio-frequency thermal ablation of the lumbar
detection and characterization of small renal vertebrae in porcine models. Radiology 2002;
masses. Radiology 1996;198(3):638-641. 224(2):452-462.
72. Rofsky NM, Bosniak MA. MR imaging in the 84. Goldberg SN, Gazelle GS, Dawson SL, Rittman
evaluation of small « or =3.0cm) renal masses. WJ, Mueller PR, Rosenthal DI. Tissue ablation
Magn Reson Imaging Clin North Am 1997;5(1): with radiofrequency using multiprobe arrays.
67-81. Acad RadiolI995;2(8):67Q-674.
73. Motzer RJ, Bander NH, Nanus DM. Renal-cell 85. Flask C, Elgort D, Wong E, Shankaranarayanan
carcinoma. N Engl J Med 1996;335(12):865-875. A, Lewin J, Wendt M. A method for fast 3D
74. Lerner SE, Hawkins CA, Blute ML, et al. Disease tracking using tuned fiducial markers and a
outcome in patients with low stage renal cell car- limited projection reconstruction FISP (LPR-
cinoma treated with nephron sparing or radical FISP) sequence. J Magn Reson Imaging 2001;
surgery. 1996. J Urol2002;167(2 pt 2):884-889. 14(5):617-627.
14
Image Guidance and Control of
Thermal Ablation
Ferenc A. Jolesz

The physical and biologic principles of localized (5,6). Even a noninvasive, targeted heat depo-
high-temperature thermal therapy are well sition method, high-intensity focused ultra-
understood. If the targeted tissue volume is sound (HIFU), has reached technical maturity
heated beyond 57° to 60°C, the threshold for (7,8). This improvement and refinement of heat
protein denaturation, then coagulation necrosis delivery devices are the primary reasons for the
occurs. This type of thermal treatment results in current emergence of minimally invasive abla-
irreversible cell damage in both normal and tion therapies. There are, however, additional
neoplastic tissues. Since heat energy deposited obstacles that hinder a broader acceptance and
above this critical level is not selective, thermal further evolution of ablation methods. Among
ablation is more comparable to surgery than to those the most important is the lack of appro-
the more selective hyperthermia. In the case of priate "closed-loop" control of efficient and
cryoablation, the underlying physical and bio- safe energy deposition.
logic principles are less well understood; never- Heat or cold transfer into deep-seated
theless multiple freezings at a relatively low organs requires the introduction of the heat-
temperature also result in cell death. conducting probes into the target volume
Despite the straightforwardness of the ther- without significant collateral tissue damage.
mal ablation approach, the clinical implemen- Although most of the ablation methods use a
tation of various ablative techniques has been relatively small-diameter probe, and therefore
challenging. There are multiple reasons for the they are, in general, minimally invasive, these
slow and gradual translation of promising probes impose major limitations on the size and
experimental results into clinical practice. Ini- shape of thermally induced tissue injury. The
tially, these were mostly technical difficulties thermal diffusion originating from these probes
because of the lack of small, percutaneously is influenced by the physical properties of the
introducible energy-delivery probes that ful- tissue, and the resulting geometry may not
fill the requirements of a minimally invasive exactly correspond to the shape of the treated
intervention. tumor. This mismatch can be corrected only if
The use of optical fibers for interstitial laser the size of the treated tissue volume increases
therapy (ILT) and the medical application of to the extent that the irregular tumor outline
radiofrequency (RF) and microwave devices fits into the larger geometrically fixed object
significantly advanced the ablation field by boundary. This may result in the destruction of
allowing percutaneous treatment (1-4). Simi- a substantial normal tissue margin, which some-
larly, by substituting the larger liquid nitro- times is preferred in treating malignancies. The
gen-based probe that uses gas with much lesion size, however, in most cases cannot be
smaller devices, percutaneous cryoablation increased indefinitely because of the develop-
became a viable alternative to cryosurgery ing balance or steady state between the rate of

182
14. Image Guidance and Control of Thermal Ablation 183

energy deposition and absorption. In most planning; this information must be updated by
cases, tissue perfusion and the presence of intraprocedural imaging as well The purpose of
blood flow profoundly influence and limit the this localization is to identify and confine the
size of the ablation. exact spatial extent of the diseased tissue to
The original intents of thermal ablative ther- be ablated, as well as to delineate the adjacent
apies are to confine the thermal coagulation to anatomic structures around that target volume.
the target volume, and to prevent injury to the The full understanding of anatomic relation-
surrounding normal tissues. This goal cannot be ship within the entire operational volume is
achieved unless the exact three-dimensional essential to define the potential access trajec-
(3D) extent of the target, and the spread tories through which the thermal probes can
(diffusion and convection) of the heat within be introduced with minimal tissue impairment
and outside the targeted tissue volume are (Fig. 14.1).
measured and monitored. To satisfy these Localization not only provides a diagnosti-
fundamental requirements, image-guidance cally relevant description of the target, but also
and image-guided therapy delivery control are is a target definition process used to plan and
necessary. Although thermal ablation of tumors execute the therapy. The 3D model of the target
is a well-known alternative to surgical excision, and the operational volume within which the
the control of destructive energy deposition has intervention will be carried out can be gener-
hitherto been an unresolved issue. The neces- ated from localization that comes from multi-
sity for spatial and temporal temperature modality imaging. For correct localization, not
control is most obvious for the brain, where only tumor margins are defined, but also intra-
thermal damage must be limited to the target. tumoral features are important, for example, to
It is less obvious in the liver that has plenty of differentiate the solid from the cystic part of a
tissue to "waste," unless the targeted volume is tumor and to distinguish necrotic tumor from
close to critical structures. It is imperative to viable neoplastic tissue. Contrast agents play a
establish the basic principles of image-guidance major role in characterizing tumor vascularity
and image-based control for thermal ablative and/or tumor boundaries. Dynamic contrast
therapies. enhancement can characterize tumor perfu-
sion, which is essential information for thermal
ablation.
The Principles of Image Localization of tumors is required to define
Guidance the ultimate target volume. The target volume
is not necessarily identical to the tumor volume.
In therapeutic procedures that require image In thermal therapies, as in surgery, when possi-
guidance, imaging methods are used for local- ble the target should be larger than the targeted
ization, targeting, monitoring, and control (9). tumor to assure a substantial "surgical" margin.
To develop and clinically implement image- This is the case with liver and breast tumors
guided ablations, it is essential to use all these during which the parenchyma beyond the
components to achieve complete treatment of detected tumor boundary can be ablated. In the
the targeted tissue volume, while minimizing case of prostate ablation, the treatment should
the associated collateral damage to surround- be confined to an anatomically defined target,
ing normal tissue. irrespective of tumor margins. Therapy plan-
ning for brain tumor ablation should consider
the presence of essential functional areas that
Localization may overlap with the tumor, and in that case,
The localization of tumor margins and the the target may be smaller than the tumor
3D definition of the targeted tumor volume volume. In the case of benign tumor ablation,
are essential requirements for image-guided such as uterine fibroids, we may elect partial
thermal therapy. The localization process is part tumor ablation so the target may represent only
of the preprocedural diagnostic workup and a fraction of the overall tumor volume.
184 F.A. Jolesz

A 8
FIGURE 14.1. Localization of the tumor is the initial in a 62-year-old man. (B) Three-dimensional (3D)
step in an image-guided thermal ablation procedure. visualization of the patient's image data provides
This takes place during the diagnosis and planning anatomic context for the tumor. Superimposed in
stages of a patient's care. The purpose is to identify this 3D space are the original axial MRI images
the location and spatial extent of the diseased tissue. (seen just below the gallbladder, perpendicular to
(A) The axial contrast-enhanced MRI reveals a the plane of the Figure). (Images provided by Stuart
solitary 2 x 2cm metastasis in segment 7 of the liver G. Silverman.)

Complete tumor destruction of most malig- possible trajectories. This targeting allows the
nant tumors is unachievable because of ill- insertion of thermal probes and matching of
defined tumor margins and the limited sensitiv- the predefined thermal volume with the target
ity of imaging. The frequent failure of thermal volume.
therapy methods is the direct consequence of Conceptually, the single-trajectory biopsy
this inaccurate target definition. Cure is easier is a key element of several therapeutic proce-
if the tumor is well defined. Conversely, in cases dures in which the insertion of needles or other
of ill-defined, diffuse, infiltrative malignant therapy-delivery probes becomes necessary.
tumor, cure is an extremely difficult task For accurate targeting, a more or less correct
and likely impossible. Tumor-seeking contrast anatomic model has to be generated from
agents and biomarkers can improve not only preprocedural or intraprocedural images. In the
the detection of tumors, but also the effective- case of preoperatively obtained image data,
ness of thermal therapies. Improving image this model should be registered to the actual
quality and spatial resolution by applying mul- patient. If there are shifts and/or deformations
timodality imaging is essential to define the full of soft tissues, the preoperative image-based 3D
extent of more invasive malignancies, such as models should be updated or corrected using
cerebral glioma and breast cancer. elastic warping algorithms or other more elab-
orate computer-based methods.
Single trajectory procedures like biopsy or
Targeting thermal probe insertion require intraproce-
Target definition is followed by the selection of dural imaging for targeting, especially in the
potential access routes. This targeting process case of mobile, deformable, or unstable soft
is essentially therapy planning that involves tissue organs. The combined or integrated local-
choosing a single trajectory from multiple ization and targeting process can be achieved
14. Image Guidance and Control of Thermal Ablation 185

either with real-time or close to real-time insufficient to monitor properly the 3D distri-
imaging methods like x-ray fluoroscopy, bution of energy. Besides, these measurements
computed tomography (CT) fluoroscopy, do not signal the irreversible cellular impair-
ultrasound (US), or intraoperative magnetic ment that is the desired end point of these
resonance imaging (MRI). Projectional or thermal ablations.
two-dimensional imaging methods, especially The various physical probes that are used
when integrated with navigational techniques, with the imaging system reflect the interaction
are suitable for interactive targeting; this between the thermal energy and the tissue. A
involves real-time planning by the interven- wide variety of imaging parameters can be used
tionalist. Using nearly real-time cross-sectional to monitor temperature. There are considerable
methods, such as US, CT, or MRI fluoroscopy, differences between imaging systems and how
interactive localization and targeting are possi- they detect the spread of thermal energy. Each
ble if the probe position can be tracked contin- imaging modality has its own limitations that
uously during the procedure by various optical are defined by their physical characteristics. The
and nonoptical sensors. Interventional MRI monitoring capabilities of imaging systems can
systems (IMRI) with multiplanar imaging capa- be compared, using certain specific attributes
bility and with navigational tools can operate such as temperature-sensitivity, temporal reso-
like US by using interactive scanning of the lution, adequacy for volumetric imaging, and
entire operational volume (Fig. 14.2) (10). ease of use during procedures. Real-time mon-
This selection of optimal trajectories for itoring devices, x-ray fluoroscopy and US are
thermal probes is the only image-guidance inexpensive methods to monitor the location
method in most currently practiced thermal and position of thermal probes, but they are
ablations. Only a limited number of application insensitive to thermal changes. Similarly, CT
sites create a 3D model of the operational has no substantial sensitivity to detect tem-
volume, target volume, and predicted thermal perature changes. Temperature-sensitive MRI
treatment volume. These 3D models should be techniques are fundamental for MRI-guided
distinctly separated from each other and should interventional procedures or for MRI-guided
be individually highlighted to facilitate the ablations. The ability of MRI to detect tem-
simulation and planning process. perature changes initiated and motivated the
Planning for thermal ablation and radiation evolution of IMRI from a relatively simple
therapy has substantial similarities. Both re- image-guidance method used for biopsies to a
quire the creation of a 3D volumetric model, complex tool to monitor and control thermal
target definition, optimization of access routes, ablations, other minimally invasive percuta-
and the avoidance of important normal neous procedures, and eventually endoscopies
anatomic structures. Radiation plans are based and open surgery (Fig. 14.3).
entirely on predictions for delayed biologic The 3D extent of the thermal injury cannot be
effects. Plans for thermal ablation also should verified without volumetric imaging. To kill
include thermal dosimetry, as well as the esti- tissue with heat, the critical dose of thermal
mation of acute and late effects. energy has to be delivered, and at least a 60°C
isotherm should envelop the entire mass. This
requires not only accurate target definition in
Monitoring 3D, but also temperature-sensitive imaging
Real-time or nearly real-time monitoring of methods that can limit the deposition of destruc-
temperature and thermally induced changes tive energy within the lesion and detect poten-
and other functional or physical parameters tial thermal energy spread to the surround-
that may be altered or modified during ablation ing normal tissue. This monitoring permits the
procedures is possible. Direct measurement or thermal ablative treatment to be highly effective
mapping of temperature distribution is possible and, at the same time, relatively safe. During
only with multiple invasive probes. The num- monitoring of thermal ablations, tissue charac-
ber of temperature-sensitive probes usually is terization still is essential. It is needed to provide
186 EA. Jolesz

A B

c D
FIGURE 14.2. Targeting is performed intraprocedu- dure. The image data are supplemented by an icon
rally. It relies on localization and planning efforts, (central, vertical dashed line) that is registered to the
as well as intraprocedural imaging. This targeting handle of the needle and assists in the targeting. The
process implements the treatment planning and oblique sagittal image (B) is orthogonal to A, and is
involves choosing a single trajectory from multiple perpendicular to the needle; the icon (crosshair) pro-
possible trajectories for the insertion of each thermal vides added trajectory information. (C) Targeting is
probe, and matching the predefined thermal volume complete with the placement of two probes (arrows)
with the target volume. Here, a kidney tumor is for effective coverage of the hyperintense tumor by
targeted for percutaneous cryoablation under prone the iceball (signal void in D) (arrows). (Images pro-
MRI guidance. (A) An oblique axial image is vided by Stuart G. Silverman.)
acquired in plane with a needle during the proce-
A–D
14. Image Guidance and Control of Thermal Ablation

E–H

Figure 14.3. (A–I) Monitoring must provide suitable spatial and temporal resolution for visualization of the
thermal event in tissue. “Real-time” imaging of a more slowly developing iceball during cryotherapy does not
require the frequency of image updates that a more rapid radiofrequency (RF) ablation does. In the top row,
three-probe cryotherapy in the liver is monitored by MRI; the ice is seen clearly as the developing region of
signal void. In the bottom row, heating with an array-type RF probe is monitored by fast thermal MRI.
187

I (Cryotherapy images provided by Stuart G. Silverman; RF images provided by Osamu Kainuma.)


188 EA. Jolesz

information as the procedure progresses and the tions have been shown. Similarly, hot spots can
original tissue undergoes phase transitions. The be present within on iceball during cryotherapy.
detection of these coagulative processes pro- Outside the target volume, these changes also
vides a relatively simple way to follow the should be monitored to assure safety and
progress of thermal ablations by detecting the prevent damage of normal tissue.
changes in tissue integrity. In a limited way,
both CT and US can be used for this "thumb-
printing" method when the ablated tissue is
Control
somewhat distinguishable from the intact one. Originally the role of image guidance during
Sequential imaging steps can follow the pro- thermal ablations was limited to the intro-
gressive tissue coagulation. duction of various probes through which the
Thermally induced phase transitions are treatment was accomplished. Although correct
best revealed on MR images; therefore, short positioning of the probes within the lesions
of thermal monitoring, this thumb-printing is extremely important, imaging should con-
method is applicable for MRI-guided ther- tinue during the energy delivery as well. The
mal interventions. The temperature-sensitivity increased role of monitoring should result
of various MR parameters [primarily inversion in the implementation of quantitative imaging,
time (TI), diffusion, and chemical shift] can based on control techniques with heat sensitive
be exploited to detect temperature changes advanced image-guidance methods.
within a critical temperature range (11-14). Imaging is important for optimal energy
Also, appropriate MR sequences can distin- delivery during thermal ablations. This image-
guish phase transitions within the thermally based control can be accomplished only if the
treated tissue volume, and can verify normal temperature-sensitivity of the imaging systems
margins that encompass the thermal lesions. and the technical capabilities of the therapy
Beyond these edges, the temperature elevation devices are matched and are functionally inte-
still is visible with MR, but is low enough to rule grated. To monitor the progressive enlargement
out cell necrosis. and reduction of the "iceball" or the expansion
The role of MRI in thermal ablative therapy of heat-treated tissue volumes, the time constant
is twofold. First, real-time MRI with tempera- of the freezing process or thermal diffusion
ture-sensitive pulse sequences can detect the should be matched with the temporal resolution
transient temperature elevations. Using this of the particular imaging modality. Within this
intraoperative information, the operator can time limit, multiplanar, multislice, or volumetric
confine the thermal treatment to the target imaging should be applied to provide a correct
tissue and can prevent the unwanted heating 3D description of the process (Fig. 14.4).
of surrounding normal tissue by discontinuing Both the temporal and spatial resolution of
the energy delivery. The second role of MRI imaging has to satisfy the requirements for the
is to delineate irreversible tissue necrosis. MR closed loop control of a thermal ablation. The
images taken intraprocedurally, but immedi- magnitude and time constants of thermal diffu-
ately after the actual treatment (i.e., delivery of sion should be seriously considered before an
a thermal dose), can verify the permanent image-guided therapy procedure is conceived,
changes within the treated tissue volume and developed, and implemented. For feasibility,
present important information about the spa- the limitations of the imaging systems, com-
tial extent of thermal ablation. For the safe and patibility between the imaging and therapy
efficient delivery of destructive energy, how- devices, and the safety features of both, should
ever, images should be taken not only after, but be taken into account before the initiation of
also during the energy transfer. the full integration process.
Monitoring of temperature changes and/or The immediate visibility of a physiologic ef-
thermally induced tissue changes is essential fect of a thermal intervention (such as perfusion
within the targeted volume to achieve complete or metabolic response to elevated temperature)
tissue kill. Heterogeneous thermal maps are is potentially achievable for sophisticated con-
useful, and cold spots within RF and laser abla- trol of the procedure. Such control assures that
14. Image Guidance and Control of Thermal Ablation 189

FIGURE 14.4. Control of ablation is an extension of provides a multiplanar presentation of intraproce-


monitoring. It is achieved by the interpretation of the dural data. In the upper half of the display screen, a
visualized effects. This requires an understanding of set of eight contiguous axial images through the
the relationship between the reversible thermal head is displayed. In the lower half of the display
effects and irreversible sequelae. Further, this must screen are 3D views of the tumor with a superim-
be done in the context of the relevant anatomy- posed colorization of the temperature (left) and
both the targeted tissues and those adjacent struc- thermal dose (right). (Image provided by Pairash
tures to be spared. The figure shows the user Saiviroonporn.)
interface of thermal therapy control software that

the temperature-induced physiologic effect dose through a probe placed within tumors.
never reaches a critical threshold of irreversible The probe's size and position, the physical
thermal damage. During thermal treatment, properties of the tissue, and the form and duty
there are significant changes in the state cycle of the energy delivery ultimately will
of blood vessels. Both vasoconstriction and limit the spatial distribution of the effects. The
vasodilatation can occur, depending on the size and shape of the thermal coagulation has
actual temperature levels. These functional to conform to the 3D configuration of the
data can be used to control energy deposition tumor. Preoperative 3D treatment planning
and adjust the level of temperature within the and dosimetry are essential to optimize energy
treated volume. delivery (2). However, in some cases this solu-
tion is impractical because it requires succes-
sive repositioning of the probes or the insertion
Discussion of multiple probes, all of which increase the
likelihood of complications.
Interstitial laser thermotherapy (ILT), RF The limitation of heat-induced, probe-
heating, and cryoablation all have important delivered thermal ablations is the presence
constraints in delivering the required energy of a large thermal gradient that results in the
190 EA. Jolesz

ambiguity of the demarcation zone between the The most appealing thermal ablation method
irreversible tissue death and the reversible is focused ultrasound (FUS). This method is
tissue damage. The transient tissue damage based on primary acoustic energy deposition
results in tissue survival and subsequent tumor and secondary thermal effects. It is noninvasive
recurrence. The unpredictable separation of and requires no probe insertion, and the appro-
treated and untreated volumes remains an priately targeted and focused high-energy US
unresolved problem. The only solution is to use beam causes no tissue damage in front of or
only the central, high-temperature zone of the beyond the target. Among thermal ablations,
large thermal gradient for tumor treatment, and the noninvasive FUS has the most potential.
to disregard the rest of the heat deposition as Introducing and integrating the temperature-
most likely ineffective. This requires tempera- sensitive MRI guidance into the targeting and
ture sensitive imaging with MRI techniques. energy delivery process revived this relatively
This real-time MRI monitoring is useful in rec- old ablation method.
ognizing the diffusion and convection of heat Magnetic resonance-guided FUS has some
within the inhomogeneous tissue environment, key advantages: It is nonincisional, so there is
where both the presence of variable micro- no entry tract of destruction. Spatial control
vasculature and the occurrence of larger blood is precise, predictable, and reproducible. The
vessels can modify the treatment plans. Flow method results in nonhemorrhagic, sharply
and tissue perfusion both can influence the rate demarcated lesions. It is instantaneous, with
and extent of energy delivery and the resulting no delayed effects. The treatment is repeatable
size of the treated tissue volumes. Constant multiple times (as opposed to radiosurgery). Its
monitoring of the procedure provides input for effects are localized and nonsystemic; after-
potential control methods that optimize treat- effects are minimal. After MRI-based localiza-
ment protocols. Since the original description tion of the target, low power energy deposition
of MRI monitoring and control of laser-tissue is used to target-a nondestructive "test" pulse.
interactions (15), MRI-guided tissue ablation The MRI-detectable small temperature eleva-
has become a clinically tested and an accepted tion within the few-millimeter diameter focal
minimally invasive treatment option. It is a rel- spot causes no permanent tissue damage, but
atively simple, straightforward method that can be located. When targeting is accomplished,
can be well adapted to the interventional MRI the power level can be increased to achieve
environment. irreversible tissue damage. This method is
Although RF treatment is basically the same now under clinical testing for the treatment of
as ILT (both based on thermal coagulation), benign and malignant breast lesions, uterine
there are differences in the resulting lesion fibroids, and brain tumors (7,8).
size. The optical laser fiber has a relatively The most serious shortcoming of thermal
small heating surface, while the RF probe can ablative treatment of malignant tumors is the
cook a larger tissue volume. The tip of the lack of precise target definition by MRI. The
optical fiber can be extremely hot, resulting in surgical concept of well-defined tumor masses
charring, which prevents the spread of the is incorrect. Instead, there are spatially dissem-
optical energy. The RF probe never reaches inated tumor cells that may spread beyond the
that high temperature (by internal cooling or reach of the thermal treatment. This is a serious
stepwise power increases), and thus the thermal handicap for both conventional and minimally
diffusion effect can be enhanced. Most RF invasive approaches. Nevertheless, if cure is not
treatments have been accomplished without anticipated, a less invasive procedure is more
temperature-sensitive imaging, and MRI has justifiable for palliation.
been used only to detect the resulting coagula- Image-guided thermal ablation requires the
tive lesion. The incompatibility of RF energy integration of therapeutic devices with imaging
deposition and MRI has been resolved, and systems. This integration is a prerequisite of
MRI-guided RF ablations have become a image-guided therapy: both location and feed-
reality (16-18). back control of the energy disposition call for a
14. Image Guidance and Control of Thermal Ablation 191

fully integrated system. We are entering a new thermal interventional therapy: comparison of
area of combined diagnostic and therapeutic TI-weighted MR sequences. J Magn Reson
applications that involve high technology. There Imaging 1994;4:65-70.
are still unresolved problems, and most impor- 7. Jolesz FA, Hynynen K. Magnetic resonance
tant is the lack of sufficient data to establish image-guided focused ultrasound surgery.
Cancer J 2002;8(suppl1):S100-12.
the clinical efficacy of the minimally invasive
8. Hynynen K, Pomeroy 0, Smith DN, et a1. MR
techniques under trial. The few MRI-guided imaging-guided focused ultrasound surgery of
thermal ablations already performed contrib- fibroadenomas in the breast: a feasibility study.
ute to the evaluation of the feasibility of these Radiology 2001;219:176-185.
techniques. 9. Jolesz FA. Image-guided procedures and the
Image-guided thermal therapy is an interdis- operating room of the future. Radiology 1997;
ciplinary effort. Not only radiologists but also 204:601-612.
other medical disciplines, computer scientists, 10. Jolesz FA, Nabavi A, Kikinis R. Integration
engineers, and physicists are contributing to of interventional MRI with computer-assisted
the development of this exciting new applica- surgery. J Magn Reson Imaging 2001;13:69-77.
11. Kuroda K, Chung AH, Hynynen K, Jolesz FA.
tion. The goal is to take advantage of advanced
Calibration of water proton chemical shift
imaging technologies-MRI, therapy sys-
with temperature for noninvasive temperature
tems, and computers-to make this exciting imaging during focused ultrasound surgery.
technology clinically more applicable, safe, and J Magn Reson Imaging 1998;8:175-181.
efficient. 12. Stollberger R, Ascher PW, Huber D, Renhart W,
Radner H, Ebner E Temperature monitoring of
interstitial thermal tissue coagulation using MR
References phase images. J Magn Reson Imaging 1997;8:
188-196.
1. Bleier AR, Jolesz FA, Cohen MS, et a1. Real time 13. Chung AH, Hynynen K. Colucci V, Oshio K,
magnetic resonance imaging of laser heat depo- Cline HE, Jolesz FA. Optimization of spoiled
sition in tissue. Magn Reson Med 1991;21: gradient-echo phase imaging for in vivo local-
132-137. ization of a focused ultrasound beam. Magn
2. Puccini S, Bar, N-K, Bublat M, Kahn T, Busse Reson Med 1996;36:745-752.
H. Simulations of thermal tissue coagulation 14. Kuroda K, Abe K. Tsutsumi S, Ishihara Y, Suzuki
and their value for the planning and monitoring Y, Sato K. Water proton magnetic resonance
of laser-induced interstitial theremotherapy spectroscopic imaging. Biomed Thermol 1994;
(UTI). Magn Reson Med 2003;49:351-362. 13:43-62.
3. Livraghi L, Goldberg SN, Lazzaroni S, Meloni F, 15. Jolesz FA, Bleier AR, Jakab P, Ruenzel PW,
Solbiati L, Gazelle GS. Small hepatocellular car- HutH K, Jako GJ. MR imaging of laser-tissue
cinoma: treatment with radio-frequency ablation interactions. Radiology 1988;168:249-253.
versus ethanol injection. Radiology 1999;210: 16. Lewin JS, Connell CF, Duerk JL, et a1. Inter-
655-661. active MRI-guided radiofrequency interstitial
4. Goldberg SN, Stein MC, Gazelle GS, Sheiman thermal ablation of abdominal tumors: clinical
RG, Kruskal JB, Clouse ME. Percutaneous trial for evaluation of safety and feasibility.
radiofrequency tissue ablation: optimization of J Magn Reson Imaging 1998;8:40--45.
pulsed-radiofrequency technique to increase 17. Steiner P, Botnar R, Dubno B, Zimmermann
coagulation necrosis. J Vasc Intervent Radiol GG, Gazelle GS, Debatin JE Radio-frequency-
1999;10:907-916. induced thermoablation: monitoring with
5. Silverman SG, Tuncali K, Adams DF, et a1. MR T1-weighted and proton-frequency-shift MR
imaging-guided percutaneous cryotherapy of imaging in an interventional 0.5 T environment.
liver tumors: initial experience. Radiology 2000; Radiology 1998;206:803-810.
217:657-664. 18. Zhang Q, Chung YC, Lewin JS, Duerk JL. A
6. Matsumoto R, Mulkern RV, Hushek SG, method for simultaneous RF ablation and MRI.
Jolesz FA. Tissue temperature monitoring for J Magn Reson Imaging 1998;8:110-114.
Section IV
Methods of Ablation
15
Percutaneous Ethanol
Injection Therapy
Tito Livraghi and Maria Franca Meloni

Percutaneous ethanol injection therapy (PElT) culation, alcohol induces necrosis of endothe-
is one of the most widely used local procedures lial cells and platelet aggregation; consequent
to treat hepatocellular carcinoma (HCe). thrombosis of small vessels is followed by
Local-regional therapies are specific ablation ischemia of the neoplastic tissue. Two char-
modalities that introduce a damaging agent acteristics of HCC favor the toxic action of
directly into the neoplastic tissue percuta- ethanol: hypervascularization and the different
neously. These techniques are capable of consistency of neoplastic versus cirrhotic tissue.
destroying the tissue chemically, such as with Since the neoplastic tissue of HCC is softer
sterile ethanol acetic acid, or thermally by than the surrounding cirrhotic tissue, ethanol
laser, microwave, or radiofrequency (1-3). diffuses within it easily and selectively.
Percutaneous ethanol injection therapy was Similarly, hypervascularization facilitates the
the first method to be proposed (4). It was uniform distribution of alcohol within the rich
conceived independently at the University of network of neoplastic vessels.
Chiba in Japan and at the Vimercate Hospital Percutaneous ethanol injection therapy is
in Milan, Italy. performed in multiple sessions in an ambula-
Initially PEIT was targeted as a palliative tory regimen (conventional technique), or,
treatment for nonsurgical patients who had when the tumor is more advanced, in a single
primary or metastatic liver lesions (4). At the session under general anesthesia with hospital-
Barcelona Conference in 2000, percutaneous ization. The former technique is generally used
techniques were classified as a curative treat- for a single HCC that is less than 4 to 5 cm in
ment for HCC (5), based on complete response diameter or for multiple HCCs with two to
obtained in selected patients (6,7). Hepato- three nodules, <3 cm in diameter. The latter
cellular carcinoma is a multicentric disease; technique is adopted for intermediate HCCs,
indeed, the first nodule is only a prelude to single or multiple, that do not occupy more than
others. Local-regional therapies can be used 30% of the hepatic volume, and with no tumor
alone or in combination with other treatment thrombus in the main portal vein branches or
modalities for long-range therapy. in the hepatic veins.

Principles and Procedure Equipment


Alcohol acts by diffusion into cells and causes Percutaneous ethanol injection therapy usually
immediate dehydration of cytoplasmic proteins is performed under ultrasound (US) guidance
with consequent coagulation necrosis. This is because real-time control allows faster execu-
followed by fibrosis. By entering into the cir- tion, precise centering of the needle in the

195
196 T. Livraghi and M.P. Meloni

target, continuous monitoring of ethanol distri-


bution, and determination of the appropriate
amount of ethanol to be injected. In some
centers computed tomography (CT) guidance
is used (8,9). The advantage of CT is greatest in
treating large tumors, in which multiple needle
placements may be necessary to ablate the
lesion completely. In these cases, CT enables
direct visualization of areas of tumor necrosis,
and identifies potentially untreated areas
within the tumor. However, CT guidance does
not permit real-time visualization of the diffu- A
sion of ethanol.
We use a commercially available US scanner
with a 3.5-MHz convex probe that has an incor-
porated, lateral needle guide. The needle is
fine-caliber (21 gauge) with a closed conical
tip and three terminal side holes (PElT,
Hakko, Tokyo, Japan). The alcohol is sterile
95%.

Conventional Technique
B
The treatment usually is performed without FIGURE 15.1. (A) Encapsulated hepatocellular carci-
sedation or local anesthesia. Patients must fast noma, 2.5 cm in size, located in segment 6 and vas-
because nausea may occur, although it is rare. cularized on the power-doppler. (B) Treatment with
After local disinfection of skin with iodized PEl in multiple sessions: the ethanol echogenic
alcohol, which also is used as a contact medium, diffuses within the neoplastic vessels shown by
the best approach is chosen. For lesions situated power-doppler.
in the right lobe of the liver, an intercostal
approach with the patient lying on the left side
often is preferable. During insertion and retrac-
tion of the needle, the patient is asked to stop if diffusion is not clearly visible. The infusion
breathing, and during the injection to breathe can be repeated after a few minutes. Significant
slowly. In the same session, 1 to 8mL of ethanol leakage of ethanol is unusual because the sur-
are injected, with one or more injections, in dif- rounding cirrhotic tissue, of greater consistency,
ferent areas depending on the distribution of forms a barrier. Good diffusion of ethanol is not
ethanol, the tolerance of the patient, the size of seen in the focal multinodular confluent variant
the tumor, the degree of vascularizaton, and the of HCC because the fibrotic component alter-
presence or absence of septations. The area per- nates with neoplastic tissue.
fused by ethanol clearly is visible as hyper- Durig the initial session of PElT, when the
echoic. The ethanol is injected slowly and the tumor is still largely viable, ethanol is easily
diffusion is controlled by real-time US moni- eliminated by the rich neoplastic circulation.
toring (Fig. 15.1). The alcohol usually diffuses Injection is continued intermittently in small
for a radius of 2 to 3 cm around the needle tip boluses until the ethanol is seen to remain
toward the periphery of the tumor. within the lesion. Since alcohol reflux can cause
The injection is stopped if there is a signifi- pain, the needle is left within the tumor for 30
cant leakage of ethanol outside the lesion, or to 40 seconds aft~r the injection, particularly
15. Percutaneous Ethanol Injection Therapy 197

in the case of superficial lesions, and then is We treat lesions <2 cm in size in three to four
removed slowly. sessions, those of 2 to 3 cm in four to six ses-
The site of the injection is chosen before each sions, and lesions of 3.5 to 5 cm in six to ten
session to ensure perfusion of the areas previ- sessions. The number of sessions is thus approx-
ously considered untreated. Color-Doppler imately twice the diameter of the lesion,
examination, particularly with echo enhancers, although it is difficult to establish a priori the
indicates the areas of viable tissue in which number of sessions or the exact quantity of
signs of vascularization persist. Each session ethanol to be administered. This therapy also
generally requires 10 to 20 minutes, and the can be used to treat tumor thrombus with
patient remains in the waiting room for 1 to portal vein (10) when the thrombus is segmen-
2 hours after the procedure. The treatment tal or subsegmental or to arrest progres-
usually is performed twice weekly. Therapy is sion toward the main branches. In this case, the
finished when perfusion of the tumor is consid- tip of the needle is positioned precisely within
ered complete, and when no vascular signs the thrombus, so the ethanol can diffuse
are detectable after US-contrast enhancement selectively.
(Fig. 15.2).

Single-Session Technique
Treatment in a single session is performed with
the patient under general anesthesia, with tra-
cheal intubation, and mechanical ventilation, to
administer an ad libitum quantity of ethanol.
The technique adapts well to operative require-
ments, for example, to obtain a period of con-
trolled apnea in the respiratory phase judged
most appropriate. The deepest portions of the
lesion are treated first, followed by the central
portion, and last, the most superficial portions
of the lesions. This sequence prevents an initial
superficial diffusion of ethanol, which, owing to
A its hyperechogenicity, would impede the guid-
ance of successive injections. Direct injection of
ethanol into hepatic veins must be avoided,
because sudden, high concentrations of alcohol
in the bloodstream may lead to prolonged
hypoxemia, followed by cardiopulmonary
collapse.
The treatment is concluded when the tumor
appears completely hyperechoic (Fig. 15.3). The
total time required for the procedure ranges
from 20 to 60 minutes, depending on the
number and size of the lesions to be treated. In
B any case, the total amount of ethanol always is
FIGURE 15.2. (A) Small hepatocellularcarcinoma less than the volume of the tumor. In the days
1.2cm in size located in the left lobe vascularized at immediately following the treatment, forma-
the USCA examination. (B) No enhancement imme- tion of gas within the lesion occurs, particularly
diately after PEl multisession treatment at contrast- when more than 50mL of ethanol have been
enhanced examination. administered.
198 T. Livraghi and M.P. Meloni

Cases of neoplastic seeding along the needle


tract have been reported, particularly in mod-
erately differentiated tumors (6,11,14).

Single-Session Treatment
Greater volumes of ethanol per session corre-
late with a higher complication rate (15). Some
cases of alcohol intoxication have been
observed, but they do not require therapy. High
levels of alcoholemia normalize within 24
hours. Abdominal pain may last 2 to 3 days, and
FIGURE 15.3. Single encapsulated hepatocellular car- is controlled with analgesic treatment. Hyper-
cinoma 6 em in size, shown by US, located in the right pyrexia for a few days is observed routinely
lobe, at the end of the treatment with single-session in patients administered more than 50mL of
PEl is quite echogenic. alcohol. During the first day of treatment, there
is an increase in transaminases, bilirubin,
d-dimer, and white blood cells, and a reduction
in fibrinogen, haptoglobin, hemoglobin, platelets,
Adverse Effects and and red blood cells. These effects usually nor-
malize within 2 weeks. The changes are due to
Complications necrosis of neoplastic tissue, and diffuse intra-
and peritumoral microthrombosis and hemoly-
Conventional Treatment sis. In our initial 111 treatments, when
Most patients complain of mild pain during or contraindications were not yet well known,
immediately after the injection, but it is usually there was one death due to bleeding from
well tolerated and does not prevent normal esophageal varices; this occurred on the third
daily activities. The pain usually is restricted to postprocedural day in a Child's class C patient
the injection site, but patients sometimes com- who had a single encapsulated HCC.
plain of pain in the right shoulder or epigastric Major complications included intraperi-
region. Patients who complain of severe pain toneal hemorrhage that required transfusion,
after the first session are given an analgesic hepatic decompensation in a Child's B patient,
before each subsequent session. transient renal insufficiency, and two cases of
No death occurred in 1623 patients treated infarction of a segment adjacent to the tumor.
with conventional PElT, collected from various Minor complications included slight bleeding
series (6). In a multicenter study that included and transitory hepatic decompensation. The
1066 patients, one death (0.09%) was reported mortality rate was 0.7%, and major complica-
due to hemoperitoneum and subsequent tions 4.6% (16,17). In another series, the mor-
hepatic coma (11). Two other anecdotal cases of tality rate was 6% (five patients): three patients
death have been reported: one due to a massive died from rupture of esophageal varices, one
area of hepatic necrosis distant from the site of patient from rupture of a subcapsular HCC,
injection of ethanol, and one to myocardial and one patient from hepatic failure (18).
infarction. In the latter case, there was no direct
correlation between the two events (12); in the
former case, the massive hepatic necrosis was Contraindications
caused by an intratumoral arterioportal shunt
that allowed ethanol to spread through the Contraindications depend on the general condi-
blood vessels (13). Major complications are tion of the patient (advanced cirrhosis or severe
rare, and occur in 1.3% to 3.2% of cases; they coagulation disorders). We do not perform
generally are treated conservatively (6,11). PElT in Child's C patients or when prothrom-
15. Percutaneous Ethanol Injection Therapy 199

bin time is significantly prolonged and platelets trast-enhanced US can be useful, but it should
<40,000/mm3• Contraindications depend on the not be used as the only test to establish follow-
tumor stage, extrahepatic metastases, the pres- up, because it is less sensitive than CT in demon-
ence of thrombosis in main portal branches, strating vascularity of small viable areas.
tumor volume >30% of the hepatic volume, or For tumor markers, we use a-fetoprotein
a diffuse form of HCG Single-session therapy is (AFP) and des-y-carboxy-prothrombin (DCP),
contraindicated in the following situations- which are often complementary. Nevertheless,
marked portal and/or pulmonary hyperten- their effect is useful only if they are initially
sion, esophageal varices at risk for bleeding, high. When the imaging shows complete re-
increased fibrinolysis, cardiac ischemia, major sponse, but AFP or DCP levels remain high, it
disorders of myocardial conduction, large sub- means that neoplastic tissue is not detected or
capsular lesions, severe coagulation disorders, is growing elsewhere. An increase always sug-
and chronic renal insufficiency. gests local recurrence or the appearance of new
lesions. Contrast-enhanced US, CT, and serum
assay of tumor markers are carried out a month
after treatment and then every 4 to 6 months
Evaluation of Therapeutic thereafter.
Efficacy
To evaluate therapeutic response, that is, to
determine whether the tumor has become com-
pletely necrotic or whether areas of neoplastic
tissue are still present, a combination of inves-
tigations and serum assays for tumor markers
is used. The follow-up tests are the same exams
as for initial staging. Although many studies are
virtually comparable, we prefer to use contrast-
enhanced US, currently with second-generation
echo enhancers (e.g., Sono Vue, Bracco, Milan,
Italy) and spiral CT with biphasic technique
(4-5 mLisec, 20 and 60 seconds after the injec-
tion of contrast medium) (Fig. 15.4).
Other examinations or biopsy are performed
only in unusual problem cases of partial versus A
complete response. If areas of tissue are still
viable but are very small beyond the present
degree of resolution, they obviously will not
be detectable. However, they will be identified
at successive examinations as they become
apparent as zones of enhancement on CT or as
they increase in volume.
Response is considered complete when CT
shows total disappearance of enhancement
within the neoplastic tissue, and is confirmed
on successive examinations (Fig. 15.5). The
absence of enhancement implies absence of B
blood flow due to necrotic and fibrotic effects. FIGURE 15.4. Hepatocellular carcinoma located in
Even with complete necrosis, the dead tissue the left lobe: contrast-enhanced computed tomogra-
occupies space and remains visible in place of phy (CT) (A), and enhanced US (B) demonstrate
the tumor, but reduced in size (Fig. 15.6). Con- hypervascular lesion (arrows).
200 T. Livraghi and M.E Meloni

A B
FIGURE 15.5. Hepatocellular carcinoma the day after ment within the lesion and a hypervascular inflam-
radiofrequency: contrast-enhanced cr (A), and matory ring (arrow).
USCA (B), demonstrate the absence of enhance-

Results parison of Child's A versus Child's C dis-


ease (p < .0001), diameter :S;3 cm versus ~4 cm
Many long-term survival curves have been pub- (p < .0002), and number :S;3 versus ~4 lesions
lished. A total of 112 patients have been treated (p < .02) showed significant differences.
at the University of Chiba-93 patients with Lencioni et al (21) treated 184 patients. A
one, 16 with two, and three with three lesions, select group of 70 patients with well-compen-
all ~3cm in diameter. Survival at 1, 3, and 5 sated cirrhosis and a single HCC 3 cm or less in
years, respectively, was 96%,72%, and 51 % for diameter had 3- and 5-year survival of 89% and
60 patients with Child's A; 90%, 62%, and 48% 63%, respectively. A multicenter Italian study
for 33 Child's B patients; and 94%,25%, and enrolled 746 patients (6). In Child's A patients
0% for 19 patients with Child's C disease (19). (n = 293), B (n = 149), or C (n = 20) with a single
Shiina et al (20) treated 50 patients with single HCC <5 cm in diameter, survival at 1,3, and 5
or multiple lesions of diameters varying from years, respectively, was 98%,79%, and 47% for
1.2 to 6 cm. Overall survival at 1, 3, and 5 years Child's A; 93%, 63%, and 29% for Child's B;
was 87%, 62%, and 43%, respectively. Com- and 64%, 12%, and 0% for Child's C. In Child's

A B
FIGURE 15.6. Hepatocellular carcinoma (arrows) treated with radiofrequency: the day after therapy (A), and
2 years later (B).
15. Percutaneous Ethanol Injection Therapy 201

A patients with multiple HCCs (n == 121), sur- 4. Alcohol therapy does not have the dis-
vival was 94%, 68%, and 36% at 1, 3, and 5 advantage of loss or damage of nonneoplastic
years, respectively. A multicenter Japanese parenchyma. In contrast, it is likely that the
study enrolled 110 Child's A patients with HCC loss of healthy tissue due to multisegmental
3 cm or less and 3 lesions or less: their 3- and resection or parenchymal damage following
5-year survival was 83% and 53%, respectively a massive transarterial chemoembolization
(22). As regards HCC >5cm in diameter, a may result in deterioration of hepatic function
study of 108 patients reported a 3-year survival and potentially terminal insufficiency (23,
of 57% in patients with encapsulated HCC 24).
measuring 5 to 8.5 cm in diameter and 42% in 5. Percutaneous ethanol injection therapy is
patients with infiltrating HCC measuring 5 to a low-risk method. In series published thus
lOcm or multiple lesions (16). far, the mortality rate has been minimal, and
In all the aforementioned patients, the main the highest complication rate is 3.2%, with
cause of death in Child's A patients was pro- most complications treated in a conservative
gression of the neoplastic disease due mainly manner. The absence of mortality with conven-
to the appearance of new lesion; in Child's C tional PElT is in marked ontrast with the peri-
patients it was hepatic insufficiency. The inci- operative mortality due to surgery, which even
dence of appearance of new lesion at 5 years in though much lower than in the past, is a factor
the group of patients from the University of to take into consideration, especially in centers
Chiba was 87%, in the Shiina group 64%, and with little experience. Centers with extensive
in the Lencioni group 78%. In our study, the surgical experience report a mortality rate of
overall incidence was 87%. In patients with a 1.4% to 11 % and a complication rate as high as
single HCC, new lesions occurred in 74%, and 58% (6). In this regard, it should be remem-
in patients with multiple HCCs the incidence bered that patients selected by ultrasound
was 98%. screening are asymptomatic and that, even if
The incidence of local recurrences (i.e., due untreated, they have a life expectancy of more
to an increase in size or the appearance of than 1 year (25).
viable areas within the lesion treated with 6. Percutaneous ethanol injection therapy
PElT) was 4% in the series of Ebara et al (19) can be repeated easily when new lesions appear,
(in lesions <3 cm in diameter), 7% in the series as happens in most patients within 5 years. The
of Shiina et al (20) (in lesions <4-5 cm in diam- incidence of appearance of new lesions in sur-
eter), and 17% in our series (in lesions <5 cm gical series varies from 67% to 100% (6). In our
in diameter). Many of the recurrences were series treated with PElT, it was 65% to 98%.
treated with additional cycles of PElT. This factor of multicentricity in patients with
HCC and chronic hepatitis always should be
taken into consideration whenever a choice of
Discussion treatment is necessary. This means that the first
lesion seen at ultrasound is usually the prelude
The rationale to utilize to PElT is based on the
to others, and that surgery, even if radical for
following points:
resected lesions, is generally only palliative
1. The expansive type of HCC initially shows because of the natural history of the disease.
regional growth, so a local therapy like PElT Patients should be followed frequently to treat
can be used. lesions as soon as possible when they are still
2. Ultrasound screening of cirrhotic popula- small. An advantage of PElT is that the patient
tions identifies HCC at an early stage, when the is followed by the same physician in the diag-
tumor is still small, generally less than 5 cm. nostic as well as in the therapeutic phase. More-
3. Ethanol shows a selective diffusion in over, it is now possible with single-session PElT
HCC owing to the tumor's soft consistency with to intervene further in the course of the disease
respect to the surrounding cirrhotic tissue and in select patients who would not have benefited
its hypervascularity. from conventional PElT.
202 T. Livraghi and M.E Meloni

7. The low cost of the material and easy undergoing resection at 5 years was 54%,
execution of the procedure make it possible versus 110 patients treated with PElT, in which
to perform PElT readily. Patients generally are it was 53% (22). The substantial comparability
treated on an outpatient basis. The cost of mate- can probably be attributed to a balance
rial is not expensive: in Italy, one PElT cycle is between the advantages and disadvantages of
about $1000, the cost of an orthotopic liver the two therapies: the greater percentage of
transplantation is about $125,000, and that of complete ablation with resection favors
a partial hepatectomy is about $30,000. The surgery, while for PElT it is the absence of peri-
number of patients who will develop the dis- operative mortality, the absence of normal
ease is about 20,000 to 25,000 in Japan and tissue loss, the insufficient evaluation of prog-
about 12,000 in Italy. Thus the problem of cost nostic factors in resected patients, and the up-
is quite important (6). staging of imaging examinations.
8. The local therapeutic efficacy of PElT is The large number of patients enrolled in US-
rather high. Histopathologic studies of resected screening programs has created a demand for
lesions carried out, particularly in the initial an effective, safe, repeatable, and economic
period of this procedure demonstrated the treatment that can be made available in many
effectiveness of PElT. Shiina et al (13) demon- centers; PElT satisfies all such requisites. In the
strated complete necrosis in 16 of 23 lesions absence of randomized studies and on the basis
(69.6%),90% necrosis in six lesions, and 70% of reported results, PElT is indicated as the
necrosis in one lesion. Their study showed that treatment of choice for most patients found
viable tissue was present in small satellite nod- positive by US screening, excluding those who
ules around the main lesions, along the lesion are candidates for liver transplantation or for
margins, or in septations. surgical resection; unfortunately, the former is
9. Long-term results with PElT are satisfac- available only for few patients. Regarding the
tory. No controlled studies have reported PElT indication for surgery, the Liver Cancer Study
compared to no treatment, but the same is true Group of Japan has reported factors predictive
for surgery. It was evident from the beginning for a long-term prognosis (29). By multivariate
that PElT was an efficacious treatment with analysis, the most important predictive factors,
few risks. in decreasing order, were AFP level, tumor size,
number of lesions, age of the patient, stage of
Comparisons with no treatment are based cirrhosis, margins of resection, and portal
only on historical data. Two studies of untreated thrombosis. Univariate analysis showed capsu-
patients with comparable disease showed a 5- lar infiltration, tumor extent, and Edmondson-
year survival of 0% in 27 patients, and in a Steiner classification to be the most important
second study of 73 patients, a rate of 11 % predictive factors. In our opinion, PElT is
(19,25). A controlled study of Child's class A therefore indicated in patients who are (a) not
patients with HCC less than 4 cm in diameter candidates for liver transplantation; (b) not
confirmed the validity of PElT compared to no resectable; (c) resectable, but presenting with
treatment, as survival at 3 years was 71 % and one of the negative prognostic factors of the
21 %, respectively (27). aforementioned multivariate analysis (high
In a comparative study based on historical AFp, age over 70 years, multinodularity, Child's
data, the mean overall survival at 5 years for class B disease, vascular infiltration); or (d)
628 patients with lesions less than or equal to resectable with only enucleation, since such
5 cm in diameter and compensated cirrhosis surgery does not remove eventual peritumoral
treated with PElT was 48%, whereas that of satellites (which is also a limitation of PElT).
1272 patients undergoing surgical resection Patients undergoing enucleation, in addition to
with a similar presentation was 49% (6). This having a significantly shorter survival than
comparability was confirmed in a cohort study those who undergo segmentectomy or subseg-
(28), and, more recently, by a multicenter mentectomy, also have a shorter survival than
Japanese study, in which survival of 445 patients that obtained with PElT (30).
15. Percutaneous Ethanol Injection Therapy 203

References 13. Boucher E, Carsin A, Raoul JL, Marchetti C,


Joram F, Kerbrat P. Massive hepatic necrosis sec-
1. Seki T, Wakabayashi M, Nakagawa T, et al. Per- ondary to treatment of hepatocellular carcinoma
cutaneous microwave coagulation therapy for by percutaneous ethanol injection. Gastroen-
solitary metastatic liver tumors from colorectal terol Clin BioI 1998;22:559-561.
cancer: a pilot clinical study. Am J Gastroenterol 14. Ishii H, Okada S, Okusaka T, et al. Needle tract
1999;94:322-327. implantation of hepatocellular carcinoma after
2. Amin Z, Donald JJ, Masters A, Kant R, Bown percutaneous ethanol injection. Cancer 1998;82:
SG, Lees WR. Hepatic metastases: interstitial 1638-1642.
laser photocoagulation with real-time US moni- 15. Giorgio A, Tarantino L, Francica G, et al. One-
toring and dynamic CT evaluation of treatment. shot percutaneous ethanol injection of liver
Radiology 1993;187:339-347. tumors under general anesthesia: preliminary
3. Rossi S, Buscarini E, Garbagnati F, et al. Percu- data on efficacy and complications. Cardiovasc
taneous treatment of small hepatic tumors by an Intervent Radiol 1996;19:27-31.
expandable RF needle electrode. AJR 1998;170: 16. Giorgio A, Tarantino L, Francica G, et al. One-
1015-1022. shot percutaneous ethanol injection of liver
4. Livraghi T, Festi D, Monti F, Salmi A, Vettori C. tumors under general anesthesia. CVIR
US-guided percutaneous alcohol injection of 1996;19:27-31.
small hepatic and abdominal tumors. Radiology 17. Meloni F, Lazzaroni S, Livraghi T. Percutaneous
1986;161:309-312. ethanol injection: single session treatment. Eur J
5. Bruix J, Sherman M, LIovet JM, et al. Clinical Ultrasound 2001;13:107-115.
management of hepatocellular carcinoma. 18. Giorgio A, Tarantino L, Mariniello N, et al.
Conclusions of the Barcelona-2000 EASL Percutaneous ethanol injection under general
Conference. J Hepatol 2000. anesthesia for hepatocellular carcinoma: 3 year
6. Livraghi T, Giorgio A, Marin G, et al. Hepato- survival in 112 patients. Eur J Ultrasound 1998;
cellular carcinoma and cirrhosis in 746 patients: 8:201-208.
long-term results of percutaneous ethanol injec- 19. Ebara M, Otho M, Sugiura N, Okuda K, Kondo
tion. Radiology 1995;197:101-108. K. Percutaneous ethanol injection for the treat-
7. Livraghi T, Benedini V, Lazzaroni S, Meloni F, ment of small hepatocellular carcinoma: study
Torzilli G, Vettori C. Long-term results of single of 95 patients. J Gastroenterol Hepatol 1990;5:
session PEl in patients with large hepatocellular 616-626.
carcinoma. Cancer 1998;83:48-57. 20. Shiina S, Tagawa K, Niwa Y, et al. Percutaneous
8. Redvanly RD, Chezmar JL. Percutaneous ethanol injection therapy for hepatocellular car-
ethanol ablation therapy of malignant hepatic cinoma: results in 146 patients. AJR 1993;160:
tumors using CT guidance. Semin Intervent 1023-1028.
RadioI1993;10:82-87. 21. Lencioni R, Pinto F. Armillotta N, et al. Long-
9. Lee MJ. Mueller PR, Dawson SL, et al. Per- term results of percutaneous ethanol injection
cutaneous ethanol injection for the treatment for hepatocellular carcinoma in cirrhosis: a
of hepatic tumors: indications, mechanism of European experience. Eur Radiol 1997;7:514-
action, technique, and efficacy. AJR 1995;164: 519.
215-220. 22. Ryu M, Shimamura Y, Kinoshita T, et al. Thera-
10. Livraghi T, Grigioni W, Mazziotti A, Sangalli G, peutic results of resection, TAE and PEl in 3225
Vettori C. Percutaneous ethanol injection of patients with hepatocellular carcinoma: a retro-
portal thrombosis in hepatocellular carcinoma: a spective multicenter study. Jpn J Clin Oncol
new possible treatment. Tumori 1990;76:394- 1997;27:251-257.
397. 23. Yamashita Y, Torashima M, Ognuni T, et al.
11. Di Stasi M, Buscarini L, Livraghi T, et al. Per- Liver parenchymal changes after transcatheter
cutaneous ethanol injection in the treatment of arterial embolization therapy for hepatoma.
hepatocellular carcinoma: a multicentric survey CT evaluation. Abdom Imaging 1993;18:352-
of evaluation practices and complication rates. 356.
Scand J Gastroenterol 1997;32:1168-1173. 24. Groupe d'Etude et de Traitment du Carcinome
12. Taavitsainen M, Vehmas T, Kauppila R. Fatal Hepatocellulare. A comparison of lipiodol
liver necrosis following percutaneous ethanol chemoembolization and conservative treatment
injection for hepatocellular carcinoma. Abdom for unresectable hepatocellular carcinoma.
Imaging 1993;18:357-359. N Engl J Med 1995;332:1256--1261.
204 T. Livraghi and M.E Meloni

25. Livraghi T, Bolondi L, Buscarini L, et al. No 28. Castells, A, Bruix J, Bru C. Treatment of small
treatment, resection and ethanol injection in hepatocellular carcinoma in cirrhotic patients: a
hepatocellular carcinoma: a retrospective analy- cohort study comparing surgical resection and
sis of survival in 391 patients with cirrhosis. percutaneous ethanol injection. Hepatology
J Hepatol 1995;22:522-526. 1993;18:1121-1126.
26. Shiina S, Tagawa K, Unuma T. Percutaneous 29. The Liver Cancer Study Group of Japan. Pre-
ethanol injection therapy for hepatocellular car- dictive factors for long-term prognosis after
cinoma: a histopathologic study. Cancer 1991;68: partial hepatectomy for patients with hepatocel-
1524-1530. lular carcinoma. Cancer 1994;74:2772-2780.
27. Orlando A, Cottone M, Virdone R, et al. Treat- 30. Takayama T, Makuuchi M. Surgical resection.
ment of small hepatocellular carcinoma asso- In: Livraghi T, Makuuchi M, Buscarini L, eds.
ciated with cirrhosis by percutaneous ethanol Diagnosis and Treatment of Hepatocellular
injection. Scand J Gastroenterol 1997;32:598- Carcinoma. London: Greenwich Medical Media,
603. 1997:279-286.
16
Radiofrequency Ablation
Riccardo Lencioni and Laura Crocetti

The goal of radiofrequency (RF) ablation is toxic temperatures. Thus, an essential objective
to induce thermal injury to the tissue through of ablative therapy is achievement and mainte-
electromagnetic energy deposition. The term nance of a 50 to lOOoe temperature through-
0

radiofrequency refers to the alternating electric out the entire target volume for at least 4 to 6
current that oscillates in the range of high minutes. However, the relatively slow thermal
frequency (200-1200kHz). In RF ablation, conduction from the electrode surface through
the patient is part of a closed-loop circuit that the tissues increases the duration of application
includes an RF generator, an electrode needle, to 10 to 30 minutes. On the other hand, the
and a large dispersive electrode (ground pads). tissue temperature should not be increased
An alternating electric field is created within over these values to avoid carbonization
the tissue of the patient. Because of the rela- around the tip of the electrode from excessive
tively high electrical resistance of tissue in heating (1,2).
comparison with the metal electrodes, there is Another important factor that affects the
marked agitation of the ions present in the success of RF thermal ablation is the ability to
target tissue that surrounds the electrode, since ablate all viable tumor tissue and an adequate
the tissue ions attempt to follow the changes in tumor-free margin. The most important
direction of alternating electric current. The difference between surgical resection and RF
agitation results in frictional heat around the ablation of hepatic tumors is the surgeon's
electrode. The discrepancy between the small insistence on a l-cm-wide tumor-free zone
surface area of the needle electrode and the along the resection margin. To achieve rates of
large area of the ground pads causes the gen- local tumor recurrence with RF ablation that
erated heat to be focused and concentrated are comparable to those obtained with hepatic
around the needle electrode (1). resection, physicians should produce a 360-
The thermal damage caused by RF heating degree, l-cm-thick tumor-free margin around
is dependent on both the tissue temperature each tumor (3) (Fig. 16.1). This cuff is necessary
that is achieved and the duration of heating. to assure that all microscopic invasion around
Heating of tissue at 50 to 55°e for 4 to 6
0
the periphery of a tumor has been eradicated.
minutes produces irreversible cellular damage. Thus, the target diameter of an ablation must
At temperatures between 60 and lOOoe nearly
0
be ideally 2cm larger than the diameter of the
immediate coagulation of tissue is induced, tumor that undergoes treatment (4,5). Eradica-
with irreversible damage to mitochondrial and tion of a tumor, therefore, can be achieved with
cytosolic enzymes of the cells. At more than a single ablation if the diameter of the tumor is
1000 to 1100 e, tissue vaporizes and carbonizes. 2cm less than the diameter of tissue ablated.
For adequate destruction of tumor tissue, the For example, a 5-cm ablation device can be
entire target volume must be subjected to cyto- used to treat a 3-cm-diameter tumor. Other-

205
206 R. Lencioni and L. Crocetti

convection by means of blood circulation


(5). These processes, together with the rapid
decrease in heating at a distance from the elec-
trode, limit the extent of induced coagulation
from a single, unmodified monopolar electrode
to no greater than 1.6cm in diameter (7,8).
Therefore, most investigators have devoted
their attention to strategies that increase the
energy deposited into the tissues. Several cor-
porations have manufactured new RF-ablation
devices based on technologic advances that
increase heating efficacy. To accomplish this
increase, the RF output of all commercially
available generators has been increased to
150 to 200 W, which potentially may increase
the intensity of the RF current deposited in
the tissues. Expandable, multineedle electrodes
FIGURE 16.1. Computer art demonstrating concept permit the deposition of this energy over a
of 360-degree, l-cm margin around a tumor. larger volume. In addition, this design decreases
the distance between the tissue and the elec-
trode, thereby ensuring more uniform heating
wise, multiple overlapping ablations can be that relies less on heat conduction over a large
performed (1,4). distance (9).
Heat efficacy is defined as the difference The commercially available devices also
between the amount of heat produced and were developed strategically to monitor the
the amount of heat lost. Therefore, effective ablation process so that high-temperature coag-
ablation can be achieved by optimizing heat ulation can occur without exceeding a 110°C
production and minimizing heat loss within the maximum temperature threshold. One device
area to be ablated. The relationship between (RITA Medical Systems) relies on direct tem-
these factors has been characterized as the perature measurement throughout the tissue to
"bioheat equation". The bioheat equation gov- prevent any electrode in a multi-tined configu-
erning RF-induced heat transfer through tissue ration from exceeding 110°C. The two other
has been previously described by Pennes (6). commercially available devices (Radionics and
The equation has been simplified to a first Radiotherapeutics) rely on an electrical mea-
approximation by Goldberg and colleagues (7): surement of tissue impedance to determine that
tissue boiling is taking place. These impedance
Coagulation == Energy Deposited rises can be detected by the generator, which
x Local Tissue Interactions then can reduce the current output to a preset
- Heat Lost level.
One of the manufacturers markets a system
Heat production is correlated with the with retractable needle electrodes (Starburst
intensity and duration of the RF energy that is XL and Starburst XLi; RITA Medical
deposited. Tissues cannot be heated to greater Systems). The needle electrodes consist of a 14-
than 100° to 110°C without vaporizing, and gauge insulated outer needle that houses nine
this process produces significant gas that both retractable curved electrodes of various
serves as an insulator and retards the ability to lengths. When the electrodes are extended, the
effectively establish an RF field. On the other device assumes the approximate configuration
hand, heat conduction or diffusion usually is of a Christmas tree, with the length and diam-
explained as a factor of heat loss in regard to eter of the electrode clusters measuring a
the electrode tip. Heat is lost mainly through maximum of 5 cm. Five of the electrodes are
16. Radiofrequency Ablation 207

hollow and contain thermocouples in their tips Alternate strategies to increase the energy
that are used to measure the temperature of the deposited from a single RF electrode also
adjacent tissue. The alternating electric current have been developed. A third manufacturer
generator is available in a 150- or 200-W model. (Radionics) uses an internally cooled electrode
To perform a typical ablation, two grounding designed to minimize carbonization and gas
pads are placed on the patient's thighs. The tip formation around the needle tip by eliminating
of the needle (with retracted electrodes) is excess heat near the electrode. The needle has
advanced to the proximal edge of the lesion, an exposed tip of variable length. The tip of the
and the electrodes are deployed approximately needle is closed and contains a thermocouple
step by step from 2cm to the desired expansion. to record the temperature of the adjacent
The generator is turned on, and can run manu- tissue. The shaft of the needle has two internal
ally or by an automated program. The temper- channels to allow the needle to be perfused
ature at the tips of the electrodes are with chilled water. Conceptually, the cooling is
controlled, and the peak power is maintained believed to prevent desiccation and charring
until the temperature exceeds the preselected around the needle tip. To increase the size of
target temperature (typically between 90° the ablation, the company placed three of the
and 100°e). After the target temperature is cooled needles in a parallel triangular cluster
achieved, the curved electrodes can be with a common hub. This device produces a sig-
advanced as previously described to full nificantly larger ablation than does a single
deployment, while maintaining the target tem- cooled needle (11-13). Pulsing of RF energy
perature. When the electrodes are fully (i.e., alternation of very high RF current for
deployed, the program maintains the target several seconds, followed by minimal RF depo-
temperature by regulating the wattage. As the sition for a defined period) also has been
tissue begins to desiccate, the amount of power described as a method to increase current
needed to maintain the target temperature deposition.
decreases. At the end of the procedure, when Despite technologic advances and electrode
the generator runs off, a "cool-down cycle" is modifications that have effectively increased RF
automatically performed. After retracting the energy deposition and tissue heating,
hooks, coagulation of the needle tract can be inadequate coagulation is a clinical problem in
done (tract ablation) maintaining temperature some circumstances, due not only to the
above 70°C. large size of lesions. Key culprits implicated in
Another RF-ablation device (LeVeen inadequate coagulation include the other two
Needle Electrode; Radiotherapeutics) has elements of the bioheat equation: (a) hetero-
retractable curved electrodes and an insulated geneity of tissue composition, by which differ-
14-gauge outer needle that houses 10 solid ences in tumor tissue density, including fibrosis
retractable curved electrodes that, when and calcification, alter electrical and thermal
deployed, assume the configuration of an conductance; and (b) blood flow, by which
umbrella (10). The electrodes are manufactured perfusion-mediated tissue cooling (vascular
in different lengths. Two ground pads are used flow) reduces the extent of thermally induced
with the device; both are placed on the patient's coagulation (Fig. 16.2). These limitations have
thighs. For use, the tip of the needle is advanced led investigators to study adjuvant therapies
to the target tissue and the curved electrodes that modify the underlying tumor physiologic
are deployed to full extension. The generator is characteristics in an attempt to improve RF
switched on, and energy is administered until a thermal ablation, either in conjunction with, or
rapid rise in impedance occurs. The ablation as an alternative to, multiple ablations of a given
algorithm is based on tissue impedance, rather tumor. These adjuvant therapies can be classi-
than tissue temperature. The impedance of fied, on the basis of the bioheat equa-
the tissue increases as the tissue desiccates. It is tion, as: (a) strategies that permit an increase
believed that an ablation is successful if the in the overall deposition of energy through an
device impedes out. alteration in tissue electrical conductivity;
208 R. Lencioni and L. Crocetti

A
FIGURE 16.2. A 14-year-old girl with gastrointestinal demonstrates an 18-mm ablation (arrowhead) of the
stromal tumor metastatic to the liver. (A) Contrast- tumor. The ablation was performed with the Radion-
enhanced computed tomography (CT) scan through ics cluster electrode and a 12-minute automated
the caudal aspect of the right lobe of the liver shows ablation cycle. Note the relatively small ablation,
a 1-cm tumor (arrow) in segment 6. (B) Contrast- which was limited in size due to hyperdynamic blood
enhanced CT scan of the liver performed immedi- flow through the liver.
ately after percutaneous radiofrequency ablation

(b) strategies that improve heat retention within ductivity, because of the difficulty of achieving
the tissue; and (c) strategies that decrease the uniform fluid diffusion and distribution. Irreg-
tolerance of tumor tissue to heat (7). ularly shaped areas of coagulation have been
For a given RF current, power deposition is observed previously with RF during simultane-
strongly dependent on local electrical conduc- ous saline injection, and are attributed to
tivity. Intratumoral injection of saline solution nonuniform saline distribution (15). Saline-
prior to, or during the application of, RF enhanced ablation may lead to distortion of the
current alters tissue conductivity, thereby lesion shape at coagulation owing to the spread
allowing greater deposition of RF current of hot fluid along the path of least resistance
and increased tissue heating and coagulation. and thus causing thermal damage off-target, as
Experimental findings demonstrate that abla- has been demonstrated in cow udder tissue in
tive temperatures can be generated farther vitro (16,17). Goldberg et al (14) have observed
from an RF electrode by increasing tissue elec- this phenomenon in tissue samples in which
trical conductivity with NaCI solution injection. large volumes of saline were injected (25 mL).
This method permits greater energy deposi- The administration of only relatively small
tion without inducing tissue boiling, which, in volumes of fluid may potentially minimize the
effect, flattens the heat distribution curve (14). issue of NaCl solution diffusion over larger
However, because both volume and concentra- volumes by obtaining maximal tissue heating
tion of saline solutions influence tissue heating and coagulation.
and the coagulation diameter in a nonlinear Perfusion-mediated tissue cooling reduces
fashion, optimal parameters for injection of the extent of coagulation produced by thermal
saline solution must be determined for each ablation (14,18,19). Modeling of the bioheat
type of RF apparatus and for the different types equation shows that for a given tissue and
of tumor and tissue to be treated. A major power deposition, the effects of tissue blood
concern for the feasibility of adjuvant percuta- flow predominate. RF-induced coagulation is
neous injection strategies is the possibility of also more limited and variable in vivo, particu-
nonuniform alteration of tissue electrical con- larly in clinical practice than ex vivo. Coagula-
16. Radiofrequency Ablation 209

tion in vivo often is shaped by vasculature of administration) with RF ablation is


in the vicinity of the ablation. Experiments in ongoing.
which hepatic perfusion is altered by mechani- Targeting of the lesion can be performed
calor pharmacologic means during RF ablation with ultrasonography (US), computed tomog-
of normal liver tissue and tumors show that raphy (CT), or magnetic resonance imaging
blood flow is largely responsible for this reduc- (MRI), and ablation may be delivered by
tion in observed coagulation (20,21). Several means of open surgery, percutaneous access, or,
strategies to reduce blood flow during ablation in case of abdominal procedures, laparoscopy
therapy have been proposed. Total portal (23). The guidance system is chosen largely on
inflow occlusion (Pringle maneuver) has been the basis of operator preference and local
used at open laparotomy and at laparoscopy. experience. Although acoustic shadowing due
Angiographic balloon occlusion of the hepatic to nitrogen bubbles and obscuration of the US
artery can be used, but has proven helpful only image by the RF current are major disadvan-
for hypervascular tumors (21). Embolization tages, US is still accepted as a viable modality
prior to ablation with particles that occlude for guidance or monitoring during RF ablation
sinusoids such as absorbable gelatin sponge (Fig. 16.3). The recent development of CT fluo-
or iodized oil may overcome this limitation. roscopic systems may result in a larger role for
Pharmacologic modulation of blood flow and CT in the future. Similarly, the developments of
antiangiogenesis therapy are theoretically open-architecture MRI systems and MRI-
possible, but currently should be considered compatible interventional equipment have
experimental (20). resulted in increased interest in the use of this
As far as decreasing tissue resistance to modality to help guide interventional proce-
heat, combining thermal ablation with other dures (24).
therapies such as chemotherapy or chemoem- In the clinical arena, RF ablation has
bolization are considerations. The findings of been used for the treatment of a variety of
preliminary experiments suggest that adju- neoplasms, including osteoid osteoma, hepato-
vant chemotherapy may increase the ablation cellular carcinoma, renal cell carcinoma,
volume compared with RF-ablation therapy non-small-cell lung cancer (NSCLC), and
alone (22). Further research to determine hepatic, lung, cerebral, osseous, and retroperi-
optimal methods of combining chemo- toneal metastases from a variety of primary
therapeutic regimens (both agent and route tumors.

A B
FIGURE 16.3. A 48-year-old man with hepatitis B- right lobe of the liver during radiofrequency ablation
induced cirrhosis. (A) Axial sonogram demonstrates shows the hyperechoic reaction caused by micro-
a 2-cm hepatocellular carcinoma (arrow) in segment bubble formation produced by the high tempera-
8 of the liver. (B) Oblique sonogram through the tures induced during the ablation process.
210 R. Lencioni and L. Crocetti

Liver Tumor Ablation Treatment of lesions adjacent to the gall-


bladder or to the hepatic hilum entails the risk
Hepatocellular carcinoma (HCC) and metas- of thermal injury of the biliary tract. In contrast,
tases from colorectal carcinoma are the two treatment of lesions located in the vicinity of
most common malignant tumors to affect the hepatic vessels is possible, since flowing blood
liver. It has been estimated that only 5% to 15% usually "refrigerates" the vascular wall, pro-
of patients with HCC or hepatic metastases are tecting it from thermal injury. In these cases,
eligible for resection (25-27). For those patients however, the risk of incomplete ablation of the
who undergo hepatic resection, there is consid- area of neoplastic tissue adjacent to the vessel
erable postoperative morbidity, a small but real may increase because of the heat loss caused
risk of death related to the operation, significant by the vessel itself. Lesions located along the
monetary expense, and only a modest improve- surface of the liver can be considered for RF
ment in long-term prognosis. The 5-year sur- ablation, although their treatment requires
vival rate for patients undergoing resection of experienced hands and may be associated with
HCC or hepatic metastases is only 20% to 40% a higher risk of complications (Fig. 16.4).
(25-27). Most patients die from recurrent Finally, the treatment of subcapsular tumors
hepatic tumors. Although in some instances abutting other abdominal viscera poses the risk
surgery may be repeated to resect recurrent of damage to those organs. Of greatest concern
tumor, at most institutions hepatic resection is in this regard is the possibility of inducing
a "one-shot" therapy. In light of these short- thermal necrosis in an adjacent loop of bowel
comings, an effective, minimally invasive tech- (23).
nique is needed to treat these tumors, and one It has to be noticed that in HCC tumors,
that can be repeated as necessary to treat recur- RF treatment tends to produce a volume of
rent tumor. thermal necrosis that matches that of the orig-
Preoperative CT or MRI is the fundamental inallesion (Fig. 16.5). This is mainly due to the
imaging examination on which the candidacy differences in tissue impedance between
of a patient for RF ablation is based. These tumor interior and surrounding cirrhotic tissue.
imaging studies are used to determine the Radiofrequency waves do not progress easily
number and size of tumors and their relation- outside the lesion because of the reduced con-
ship to surrounding structures such as blood ductivity of cirrhotic tissue. This phenomenon
vessels, bile ducts, gallbladder, diaphragm, and has the advantages of sparing the surrounding
bowel. To be considered eligible for treatment nonneoplastic tissue and of enhancing the
by RF thermal ablation, patients must meet heat deposition within the tumor, thus increas-
some general requirements. First, as RF abla- ing the effectiveness of the procedure.
tion is a local treatment, disease ideally should However, it entails the disadvantage of lacking
be confined to the liver, without evidence of control over extracapsular daughter nodules
vascular invasion or extrahepatic metastases (29-32).
(28). In addition, the tumor must be a focal, Metastases do not have a clear lesion-to-liver
nodular-type lesion. The presence of a clear and interface like nodular-type HCCs, but tend to
easy-to-detect target for needle placement is strand into the surrounding liver parenchyma.
crucial for the outcome of treatment. The tumor Hence, if the necrosis volume produced by RF
must be either uninodular or, when multinodu- treatment is nearly equivalent to that of the
lar, preferentially with a maximum of four to visible native lesion, tumor recurrence caused
five lesions. Tumor size ideally should be by microscopic tests of tumor along the bound-
smaller than 4 to 5 cm in the greatest dimen- ary is likely to occur. Therefore, to successfully
sion. Nevertheless, larger lesions, especially ablate liver metastases, it is necessary to pro-
HCCs, also can be treated by using newer RF duce a thermal necrosis volume that exceeds
generators and RF needles, as well as by per- that of the original lesion, with ideally a 1-cm
forming RF ablation after occlusion of tumor safety margin of coagulation necrosis in
blood supply (21). the liver parenchyma all around the lesion
16. Radiofrequency Ablation 211

,,14
A .- B
FIGURE 16.4. A 52-year-old man with subcapsular mediately following ablation demonstrates the
hypervascular hepatocellular carcinoma in segment hypovascular-induced state of the ablated tumor
7. (A) Preablation contrast-enhanced CT scan (arrowhead). Note the subcapsular and subdi-
through the dome of the liver shows a 2 x 3cm aphragmatic location of the ablation, performed
hypervascular hepatocellular carcinoma (arrow). without complication.
(B) Contrast-enhanced CT scan performed im-

(Fig. 16.6). This safety margin can be achieved effect of RF treatment in the adjacent tissue
in the case of metastatic lesions, because the (29,33-35).
progression of RF waves outside the lesion is Whenever possible RF ablation is performed
not impaired by the poor conductivity of cir- percutaneously. Percutaneous treatment has
rhotic tissue as in HCC, provided that the lesion several advantages over other approaches. The
is not located in the vicinity of major vascular percutaneous approach is the least invasive,
structures. Flowing blood within the vessels acts produces minimal morbidity, requires only
as a heat sink and may limit the necrotizing conscious sedation, is relatively inexpensive,

A B
FIGURE 16.5. A 53-year-old man with hepatitis performed immediately following ablation demon-
B-induced cirrhosis with a hepatocellular carcinoma strates an avascular region of coagulative necrosis
in segment 6. (A) Contrast-enhanced CT scan (arrowhead) that closely matches the configuration
demonstrates an approximately 4-cm hepatocellular of the tumor except in the region of the watershed
carcinoma (arrow). (B) Contrast-enhanced CT scan zone where the ablation was potentiated.
212 R. Lencioni and L. Crocetti

A B
FIGURE 16.6. A 65-year-old woman with breast car- necrosis (arrowheads) at the site of the treated 1-cm
cinoma metastatic to the liver. (A) Contrast- tumors. Note that the ablations substantially exceed
enhanced CT scan through the left lobe of the liver the size of the treated tumors and clearly create a
shows two 1-cm tumors (arrows). (B) Contrast- greater than 1-cm tumor-free margin around each
enhanced CT scan performed immediately after tumor.
ablation demonstrates two large zones of coagulative

and can be repeated as necessary to treat recur- trode can be problematic. Unlike the percuta-
rent tumor. However, advocates of laparo- neous technique in which the US probe and
scopic thermal ablation of hepatic tumors claim RF needle can be moved to any position over
that the laparoscopic approach provides some the right upper quadrant to achieve the best
distinct advantages over the percutaneous angle of approach to treat a tumor, the laparo-
approach (36). With the laparoscopic tech- scopic technique is hampered by limited access
nique, the entire liver can be imaged with a through the existing laparoscopic ports. Fur-
high-frequency transducer placed directly on thermore, no needle guides currently are avail-
the surface of the liver. This technique allows able that can be attached to the laparoscopic
the visualization (and treatment) of small US transducers, and the RF needles cannot be
tumors that cannot be detected using any placed parallel to the transducers. Most com-
other imaging technique. Furthermore, with monly, the needles are placed either perpen-
laparoscopy the extent of a tumor can be staged dicular or oblique to the US transducers. The
more accurately. The identification of previ- manipulation of the needle from these angles
ously undetected surface lesions or peritoneal is technically challenging and is not easily
implants may lead to the appropriate cancella- mastered (36).
tion of the planned RF ablation. This is impor- Other investigators have published their
tant if the goal of the procedure is potential experiences with RF ablation performed via
cure. Additionally, advocates of a laparoscopic open surgical treatment (28). Disadvantages of
approach argue that they can perform a Pringle this technique include the associated mor-
maneuver (temporary occlusion of the hepatic bidity and mortality of an open procedure and
artery and portal vein) and thus enhance the general anesthetic, the added expense of the
size of a thermal ablation (36). The disadvan- procedure and associated recovery time, and
tages of a laparoscopic approach include the that the technique typically is a one-shot
added invasiveness of the procedure, with the therapy. Some of the advantages of the tech-
associated complications and added costs, and nique are the same as for the laparoscopic
the technical difficulties of the procedure. In approach-namely, the ability to scan the
particular, the placement of the needle elec- whole liver with high-frequency transducers
16. Radiofrequency Ablation 213

and to accurately stage the extent of the tumor. metastatic disease. Patients with lesions within
Additionally, the open approach allows a great 1em of the spinal cord, brain, aorta, inferior
deal of freedom in placing the US transducer vena cava, bowel, or bladder and patients with
and RF needle. Difficult lesions adjacent to impending fracture at the potential ablation
the diaphragm, bowel, or gallbladder may be site were not eligible for this study. All patients
treated better with the open technique. These experienced a decrease in pain over the course
organs can be removed or isolated from the of the follow-up period. Patients derived
mobilized liver to prevent damage during the benefit from the RF-ablation treatment within
ablation of a tumor. Finally, the blood supply to 1 week. The patients showed greater benefit
the liver may be temporarily stopped during from RF ablation with time, reporting mean
RF ablation. A Pringle maneuver, which causes decreases for pain 4 weeks after treatment. In
temporary occlusion of the portal vein and addition to providing pain relief, RF abla-
hepatic artery, decreases blood supply to both tion resulted in improved quality of life with
the targeted tumor and the adjacent hepatic decreased interference from pain in the activi-
parenchyma. The result is a larger thermal ties of daily living. The mechanism of action
injury than is possible with normal blood flow responsible for decreased pain at the metasta-
(28). tic site after RF ablation is unclear. Radiofre-
quency ablation might provide an alternative
treatment for palliation of painful metastatic
Extrahepatic Tumor Ablation lesions that are resistant to radiation and in
cases in which further radiation therapy is not
Tumors in several other organ sites have been possible because of limitations of dose to
treated with RF (2). Rosenthal et al (37,38) normal structures. It is also possible that RF
treated over 100 osteoid osteomas by using ablation will play an adjunctive role to the use
percutaneously placed conventional monopo- of radiation therapy for palliation of painful
lar electrodes. Pain, the primary clinical mani- metastatic lesions (40). Prospective comparison
festation of this lesion, was eradicated in more studies of RF ablation and radiation therapy
than 95 % of cases; 12 recurrent lesions were may be useful to help distinguish the relative
successfully re-treated with percutaneous tech- benefits of these therapies for palliation of
niques. In addition, the length of hospital stay painful metastatic lesions.
compared with that after surgical excision, pre- Additional reports have begun to appear
viously the therapy of choice, decreased from describing the use of RF ablation in other
6.8 to 2.6 days. sites and organs. Polascik et al (41) reported
Percutaneous image-guided thermal ablation their experience with saline-enhanced RF-
of metastatic neoplasms involving bone may ablation treatment of VX2 tumors that had
offer an alternative to conventional therapies been implanted beneath the renal capsule in
for pain management in terminally ill patients. rabbits. Zlotta et al (42) reported the treatment
Dupuy et al (39) reported treating 10 patients of three focal renal tumors in two patients by
with painful metastatic bone lesions by using using hooked electrodes. Extensive coagulation
CT guidance and 18-gauge internally cooled necrosis was found throughout these tumors at
electrodes. Subjective pain relief was reported subsequent nephrectomy. McGovern et al (43)
in 90% and lasted a minimum of 4 months reported the successful treatment of a single
without further treatment. Callstrom et al (40) 3-cm renal cell carcinoma (Ree) by using an
treated 12 patients with severe pain from IS-gauge internally cooled RF electrode placed
metastatic lesions involving bone that were percutaneously with US guidance.
refractory to radiation therapy, chemotherapy, In a recent series, by Gervais et al (44),42
surgery, or analgesic medicines or, alternatively, RCCs were ablated with a total of 140 overlap-
who had refused to undergo these standard ping ablations with acceptable morbidity and
therapies. Patients included in the study had no procedure-related mortality. The number of
pain resulting from no more than two sites of ablated tumors allowed statistical analysis of
214 R. Lencioni and L. Crocefti

the effect of tumor location on results of RF treated intraprocedurally with a small-bore (7-
ablation of large tumors, and the authors were Fr) percutaneous cathether that allowed the
able to achieve complete ablation of all exo- procedure to continue. Wallace et al (49) also
phytic tumors up to 5.0cm in size. Although treated patients with either primary or sec-
the mean follow-up period in this study was ondary unresectable lung malignancies. In their
short (13.2 months) with respect to the natural series of 12 patients with 16 lesions, follow-up
history of RCC, the results observed during the CT contrast densitometry demonstrated a sig-
longest follow-up period (3.5 years) were nificant drop in mean enhancement of treated
encouraging in terms of the continued long- tumors 1 to 2 months after the ablation. Com-
term evaluation of this technique. The absence plications included pneumothoraces requiring
of tumor recurrences over the time course was catheter drainage (n = 4), pleural effusion (n =
also very encouraging. Still, because many small 3), and pain (n = 2). Dupuy et al (50) treated 27
RCC tumors can be expected to grow at a rate patients, 14 with primary lung cancer and 13
of 1 to 3 mm per year, longer-term follow-up of with metastases from various primary tumors.
surviving patients to assess rates of local recur- Radiofrequency ablation was tolerated in
rence and occurrence of possible metastatic all patients, and the pneumothorax rate was
disease is needed. Small exophytic RCC tumors 14%.
(as large as 5.0cm) are the ideal tumors for In this series, residual tumor or tumor recur-
treatment with RF ablation. For patients rence at the treated site was identified in nine
with an actual or functionally solitary kidney patients after a mean follow-up of 4.4 months.
who undergo complete ablation of RCC, RF Lencioni et al (51) reported a series of 10 pa-
ablation provides a treatment option that tients with unresectable, biopsy-proven NSCLC
does not subject them to the inconvenience of 3 cm or less in greatest dimension (range,
and morbidities of dialysis. Even in the absence 1.1-3cm; mean, 2.1 ± 0.6cm) undergoing RF
of complete ablation, RF ablation may playa ablation. In this study, correct placement of the
role in palliation of disease in patients who electrode needle within the lesion was achieved
find dialysis unacceptable. This palliative role in all cases, and single-session ablation was
of RF ablation in the treatment of RCC is successfully performed. Pneumothorax was
an interesting area for further study, especially observed in four of 10 patients, two of whom
for large tumors with a central component required drainage. No other complication
(44). occurred. Pulmonary function tests did not
Dupuy et al (45) reported their experience in show any significant worsening with respect to
treating lung malignancies. Three patients with baseline. CT studies obtained 1 month after
unresectable lung tumors were treated with RF treatment showed areas of thermal injury
ablation. Zagoria et al (46) published a case encompassing the native tumor in all cases. At
report in which two lung metastatic nodules 3-month CT studies, treated areas showed
in a patient with RCC were ablated with- reduction in size with no signs of recurrence in
out major complications. Steinke et al (47) any case. Complete tumor response was con-
described a case of a pulmonary metastasis firmed in six of six patients who also were
resected after RF ablation, in which histologic assessed 6 months after the procedure. Glenn
proof of complete necrosis was obtained. et al (52) treated 10 nonsurgical patients with
VanSonnenberg et al (48) reported their initial colorectal lung metastases. Radiofrequency
clinical experience in six patients with either ablation was technically successful in 14 of 15
primary or secondary unresectable lung tumors, pulmonary tumors. Baseline tumor size was 2.6
who had exhausted radiation and chemother- ± 1.9cm (range 0.8-8.0). Computed tomogra-
apy alternatives. phy lesion size after ablation decreased from 1
In all cases, extensive necrosis, manifested as month to 3 months in all lesions with 3-month
no enhancement on the postprocedure con- follow-up. Complications requiring intervention
trast-enhanced MRI, was seen. One intraproce- occurred in three patients and included pneu-
dural pneumothorax was encountered, and mothorax (three) and pleural effusion (one).
16. Radiofrequency Ablation 215

References technique: laboratory and clinical experience


in liver metastases. Radiology 1998;209:371-
1. Rhim H, Goldberg SN, Dodd GD III, et al. 379.
Essential techniques for successful radio- 14. Goldberg S, Ahmed M, Gazelle GS, et al. Radio-
frequency thermal ablation of malignant hepatic frequency thermal ablation with NaCl solution
tumors. Radiographies 2001;21:S17-S35. injection: effect of electrical conductivity on
2. Gazelle GS, Goldberg SN, Solbiati L, Livraghi T. tissue heating and coagulation-phantom and
Tumor ablation with radiofrequency energy. porcine liver study. Radiology 2001;219:157-165.
Radiology 2000;217:633-646. 15. Livraghi T, Goldberg SN, Lazzaroni S, et al.
3. Cady B, Jenkins RL, Steele GD Jr, et al. Surgical Saline-enhanced radio-frequency tissue ablation
margin in hepatic resection for colorectal metas- in the treatment of liver metastases. Radiology
tasis: a critical and improvable determinant of 1997;202:205-210.
outcome. Ann Surg 1998;227:566-571. 16. Bohm T, Hilger I, Mueller W, Reichenbach JR,
4. Dodd GD III, Soulen M, Kane R, et al. Mini- Fleck M, Kaiser WA. Saline enhanced radiofre-
mally invasive treatment of malignant hepatic quency ablation of breast tissue: an in-vitro fea-
tumors: at the threshold of major breakthrough. sibility study. Invest Radiol2000;35:149-157.
RadioGraphies 2000;20:9-27. 17. Boehm T, Malich A, Goldberg SN, et al. Radio-
5. Patterson EJ. Scudamore CH, Owen DA, Nagy frequency tumor ablation: internally cooled
AG, Buczkowski AK. Radiofrequency ablation electrode versus saline-enhanced technique in
of porcine liver in vivo: effects of blood flow and an aggressive rabbit tumor model. Radiology
treatment time on lesion size. Ann Surg 1998; 2002;222:805-813.
227:559-565. 18. Goldberg SN, Solbiati L, Halpern EF, Gazelle
6. Pennes HH. Analysis of tissue and arterial blood GS. Variables affecting proper system grounding
temperatures in the resting human forearm. for radiofrequency ablation in an animal model.
J Appl PhysiolI948;1:93-122. J Vasc Intervent Radiol2000;11:1069-1075.
7. Goldberg SN, Gazelle GS, Mueller PRo Thermal 19. Goldberg SN, Hahn PF, Tanabe KK, et al. Per-
ablation therapy for focal malignancy: a unified cutaneous radiofrequency tissue ablation: does
approach to underlying principles, techniques, perfusion-mediated tissue cooling limit coagula-
and diagnostic imaging guidance. AJR 2000;174: tion necrosis? J Vasc Intervent Radiol 1998;9:
323-331. 101-115.
8. Goldberg SN, Gazelle GS, Halpern EF, Rittman 20. Goldberg SN, Hahn PF, Halpern E, Fogle R,
WJ. Mueller PR, Rosenthal Dr. Radiofrequency Gazelle GS. Radio-frequency tissue ablation:
tissue ablation: importance of local temperature effect of pharmacologic modulation of blood
along the electrode tip exposure in determining flow on coagulation diameter. Radiology 1998;
lesion size and shape. Acad Radiol 1996;3:212- 209:761-767.
218. 21. Rossi S, Garbagnati F, Lencioni R, et al. Percuta-
9. Rossi S, Buscarini E, Garbagnati F, et al. Percu- neous radio-frequency thermal ablation of non-
taneous treatment of small hepatic tumors by an resectable hepatocellular carcinoma after occlu-
expandable RF electrode needle. AJR 1998;170: sion of tumor blood supply. Radiology 2000;217:
1015-1022. 119-126.
10. LeVeen RF. Laser hyperthermia and radiofre- 22. Goldberg SN, Saldinger PF, Gazelle GS, et al.
quency ablation of hepatic lesions. Semin Inter- Percutaneous tumor ablation: increased necrosis
vent RadiolI997;14:313-324. with combined radio-frequency ablation and
11. Goldberg SN, Gazelle GS, Solbiati L. Rittman intratumoral doxorubicin injection in a rat
WJ, Mueller PRo Radiofrequency tissue ablation: breast tumor model. Radiology 2001;220:420-
increased lesion diameter with a perfusion elec- 427.
trode. Acad Radiol 1996;3:636-644. 23. McGahan JP, Dodd GD III. Radiofrequency
12. Goldberg SN, Gazelle GS. Radiofrequency tissue ablation of the liver: current status. AJR 2001;
ablation and techniques for increasing coagula- 176:3-16.
tion necrosis. Hepatogastroenterology 2001;48: 24. Lewin JS, Connell CF, Duerk JL, et al. Inter-
359-367. active MR-guided radiofrequency interstitial
13. Goldberg SN, Solbiati L. Hahn PF, et al. Large- thermal ablation of abdominal tumors: clinical
volume tissue ablation with radiofrequency by trial for evaluation of safety and feasibility.
using a clustered, internally cooled electrode J Magn Reson Imaging 1998;8:40-47.
216 R. Lencioni and L. Crocetti

25. Franco D, Capussotti L, Smadja C, et al. Resec- percutaneously placed electrode: a new proce-
tion of hepatocellular carcinoma: results in 72 dure. Radiology 1992;183:29-33.
European patients with cirrhosis. Gastroenterol- 38. Rosenthal DI, Springfield DS, Gebhart MC,
ogy 1990;98:733-738. Rosenberg AE, Mankin HI Osteoid osteoma:
26. Hughes KS, Rosenstein RB, Songhorabodi S, percutaneous radio-frequency ablation. Radiol-
et al. Resection of the liver for colorectal carci- ogy 1995;197:451-454.
noma metastases: a multi-institutional study of 39. Dupuy DE, Safran H, Mayo-Smith WW,
long-term survivors. Dis Colon Rectum 1988; Goldberg SN. Radiofrequency ablation of painful
31:1-4. osseous metastases. Radiology 1998;209(P):389.
27. Castells A, Bruix J, Bru C, et al. Treatment of 40. Callstrom MR, Charboneau JW, Goetz MP, et al.
small hepatocellular carcinoma in cirrhotic Painful metastases involving bone: feasibility
patients: a cohort study comparing surgical of percutaneous CT- and US-guided radio-
resection and percutaneous ethanol injection. frequency ablation. Radiology 2002;224:87-97.
Hepatology 1993;18:1121-1126. 41. Polascik TJ, Hamper U, Lee BR, et al. Ablation
28. Curley SA, Izzo F, Delrio P, et al. Radiofre- of renal tumors in a rabbit model with intersti-
quency ablation of unresectable primary and tial saline-augmented radiofrequency energy:
metastatic hepatic malignancies: results in 123 preliminary report of a new technology. Urology
patients. Ann Surg 1999;230:1-8. 1999;53:465-472.
29. Lencioni R, Cioni D, Bartolozzi C. Percutaneous 42. Zlotta AR, Wildschutz T, Raviv G, et al.
radiofrequency thermal ablation of liver malig- Radiofrequency interstitial tumor ablation
nancies: techniques, indications, imaging find- (RITA) is a possible new modality for treatment
ings, and clinical results. Abdom Imaging 2000; of renal cancer: ex vivo and in vivo experience.
26:345-360. J EndouroI1997;11:251-258.
30. Rossi S, Di Stasi M, Buscarini E, et al. Percuta- 43. McGovern FJ, Wood BJ, Goldberg SN, Mueller
neous radiofrequency interstitial thermal abla- PRo Radiofrequency ablation of renal cell carci-
tion in the treatment of small hepatocellular noma via image guided needle electrodes. J Urol
carcinoma. Cancer J Sci Am 1995;1:73-77. 1999;161:599-600.
31. Livraghi T, Goldberg SN, Lazzaroni S, et al. Small 44. Gervais DA, McGovern FJ, Arellano RS,
hepatocellular carcinoma: treatment with radio- McDougal WS, Mueller PRo Renal cell carci-
frequency ablation versus ethanol injection. noma: clinical experience and technical success
Radiology 1999;210:655-661. with radio-frequency ablation of 42 tumors.
32. Lencioni R, Allgaier HP, Cioni D, et al. A ran- Radiology 2003;226:417-424.
domized comparison of radiofrequency thermal 45. Dupuy DE, Zagoria RJ, Akerley W, Mayo-Smith
ablation and percutaneous ethanol injection for WW, Kavanaugh PV, Safran H. Percutaneous
the treatment of small hepatocellular carcinoma RF ablation of malignancies in the lung. AJR
in cirrhosis. Radiology 2003;228:235-240. 2000;174:57-60.
33. Lencioni R, Goletti 0, Armillotta N, et al. Radio- 46. Zagoria RJ, Chen MY, Kavanagh PV, Torti FM.
frequency thermal ablation of liver metastases Radio-frequency ablation of lung metastases
with a cooled-tip electrode needle: results of a from renal cell carcinoma. J Urol 2001;166:
pilot clinical trial. Eur RadioI1998;8:1205-1211. 1827-1828.
34. Solbiati L, Goldberg SN, Ierace T, et al. Hepatic 47. Steinke K, Habicht J, Thomsen S, Soler M, Jacob
metastases: percutaneous radiofrequency abla- LA. CT-guided radiofrequency ablation of a
tion with cooled-tip electrodes. Radiology 1997; pulmonary metastasis followed by surgical resec-
205:367-373. tion. Cardiovasc Intervent Radiol 2002;25:543-
35. Gillams AR, Lees WR. Survival after percuta- 546.
neous, image-guided, thermal ablation of hepatic 48. VanSonnenberg E, Shankar S, Tuncali KT,
metastases from colorectal cancer. Dis Colon Silverman SG, Morrison PR, Jaklitsch M. Initial
Rectum 2000;43:656-661. clinical experience with RF ablation of lung
36. Siperstein AE, Rogers SJ, Hansen PD, tumors. Radiology 2002;225(P):291.
Gitomirsky A. Laparoscopic thermal ablation 49. Wallace AB, Suh RD, Sheehan RE, Heinze SB,
of hepatic neuroendocrine tumor metastases. Goldin JG. Contrast nodule densitometry to
Surgery 1997;122:1147-1155. assess the efficacy of radiofrequency ablation
37. Rosenthal DI, Alexander A, Rosenberg AE, of pulmonary malignancies. Radiology 2002;
Winfield D. Ablation of osteoid osteomas with a 225(P):292.
16. Radiofrequency Ablation 217

50. Dupuy DE, Mayo-Smith WW, Di Petrillo T, frequency thermal ablation of small unresect-
Ridlen MS, Murphy BL, Cronan JJ. Clinical able non-small cell lung cancer. Radiology 2003;
experience of pulmonary radiofrequency abla- 226:38A.
tion in 27 patients. Radiology 2001;221(P): 52. Glenn DW, Clark W, Morris DL, King J, Zhao 1.
389. Percutaneous radiofrequency ablation of col-
51. Lencioni R, Crocetti L, Cioni R, Mussi A, orectal pulmonary metastases. Radiology 2001;
Angeletti CA, Bartolozzi C. Percutaneous radio- 221(P):315.
17a
Microwave Coagulation Therapy for
Liver Tumors
Toshihito Seki

As treatment for hepatocellular carcinoma capsule. Often viable cancer cells persist in
(HCC), surgical resection, transcatheter arter- the extra- or intracapsular area in surgically
ial embolization (TAE), and ultrasound (US)- resected or autopsy specimens after PElT.
guided local treatment have been performed These viable cells have the potential to cause
alone or in combination. Surgical resection is local recurrence, as well as intrahepatic or
not a viable option for all patients due to poor distant metastases.
liver function induced by hepatic cirrhosis, and To address these problems, we developed US-
TAE sometimes is ineffective because of angio- guided percutaneous microwave coagulation
neogenesis in small HCCs. For these reasons, therapy (PMCT) as a local percutaneous treat-
US-guided percutaneous local treatment has ment to induce complete tumor necrosis (1,2).
been adopted independently or in combination Regardless of the presence of a capsule, micro-
with TAE. wave irradiation is most reliable to induce tissue
Percutaneous ethanol injection therapy coagulation if the tip of the electrode is located
(PElT) was widely performed for patients in the target lesion.
with small HCCs as a local treatment in Japan Initially, treating HCC, microwave coagula-
when surgery was not indicated. Because PElT tion was used in hepatic resection during open
was tolerated by most patients, it could be surgery for tissue coagulation and hemostasis
carried out easily in those with hepatic cirrho- (3). Several studies have reported excellent
sis. However, PElT was evaluated histopatho- results (4,5). For percutaneous treatment, we
logically and clinically, and the injected ethanol could not use the electrode designed for
did not always cause complete necrosis of the hepatic resection because the shaft of the
tumor. electrode for surgical use is too thick (1 cm in
It is common to see HCCs surrounded by diameter). For percutaneous use of microwave
a fibrous capsule. However, formation of coagulation, it was necessary to develop a thin
a fibrous capsule around the tumor often is electrode. Furthermore, it had to be designed
incomplete in small HCCs. In such instances, so that this electrode caused tissue coagulation
injected ethanol may spread to the noncancer- at the tip of the electrode, but did not burn
ous peripheral tissue or may flow into large the skin. We designed a thin electrode with
vessels around the tumor instead of remaining decreased heat conduction through the shaft of
within the tumor, causing failure of tumor the electrode.
necrosis. Even when a fibrous capsule exists, but In this chapter, both the mechanisms of micro-
the encapsulated area or its periphery has been wave coagulation and the technique of PMCT
infiltrated by tumor cells, the injected ethanol for HCCs are elucidated. Clinical data using
may be blocked by the capsule and fail to reach PMCT for HCCs including complications of
tumor cells that have spread outside the PMCT are discussed as well.

218
17a. Microwave Coagulation Therapy for Liver Tumors 219

Microwave Tissue Coagulation


In tissue, microwave irradiation from a
monopolar antenna causes water molecules in
the dielectric substance to vibrate dramatically
at a frequency of 2450 MHz. This generates fric-
tional heat in the water molecules and leads to
thermal coagulation of tissue (5). Microwaves
with a frequency of 2450 ± 50 MHz and a wave-
length of 12-cm are recognized internationally
as an industrial, scientific, and medical (ISM)
band.
FIGURE17a.1. Microwave generator: Microtaze:
AZM-520, AZWELL, Osaka, Japan.
Microwave Coagulation System
The extent of the coagulation achieved by
microwave irradiation is related to the amount Microwave Electrode
of electric power and the irradiation time.
To achieve tissue coagulation of not only the The structure of the electrode (AZWELL)
tumor but also neighboring noncancerous from the tip to the other end consists of an
tissue, it is necessary to apply high energy effi- antenna lOmm long made of stainless steel, an
ciently to heat the tissue. For this purpose, the insulator 2mm long made of polytetrafluorethyl-
microwave irradiation power (in watts) must ene (PTFE), and then a coaxial system 238mm
be sufficiently high and the electrodes must be long, of which the inner conductive rod is made
able to withstand the conditions. As for irradi- of silver-plated tool steel and is electrically con-
ation time, long irradiation can induce exten- nected to the antenna. The outer conductor of
sive tissue coagulation, but is stressful for the coaxial construction is made of nickel-
patients. Therefore, irradiation time should be plated brass and this is the electrode. The
shortened as much as possible. electrode measures 2.0 mm in diameter and
25 cm in length. To prevent adhesion of the
coagulated tissue to the electrode, the surfaces
of the center and outer conductors are coated
Microwave Generator with PTFE (Fig. 17a.2).
The microwave is generated by a magnetron in It is known from the specific absorption rate
a microwave generator (Microtaze: AZM-520, (SAR) distribution that heating is limited to the
AZWELL, Osaka, Japan) (Fig. 17a.l). The area adjacent to the antenna. The electrode
electric energy of the microwave is supplied generates an electromagnetic field that sur-
to a high-frequency coaxial cable with an rounds the insulator, and the heated area of the
impedance of 50 ohms via an isolated sympa- tissue is limited to and focused on the area adja-
thizer cavity in the generator, and then con- cent to the tip of the electrode.
ducted to an electrode attached to the other
end of the cable for irradiation from the tip to
the tissue. For easy manipulation, we use a
Power Output Assessment
coaxial cable, 2.5 m long with a diameter of In vitro evaluation of coagulation capability
5.7mm. To avoid unintended and unexpected was performed using resected pig liver. With
irradiation, the generator is equipped with a irradiation at 80 W for 5 minutes, the coagu-
fail-safe system so that the microwave cannot lated area was elliptical, with maximal and
be irradiated unless both the inner and outer minimal diameters of 3.5 cm and 2.5 cm, respec-
electrodes completely contact the tissue. tively (Fig. 17a.3). At this power and duration,
220 T. Seki

FIGURE 17a.2. Microwave electrode.

the temperature of the shaft of the electrode determined. Local anesthesia is induced along
did not exceed 46°C. If the power and duration the puncture line by introducing 0.5% lidocaine
of irradiation exceeded 80W and 5 minutes, from the insertion point at the skin to the
respectively, the temperature of the shaft was peritoneum.
over 50°C. At this temperature, the skin may 2. The insertion point of the skin is incised.
burn where the electrode has been inserted. 3. Under ultrasonic guidance, a 13-gauge
guide needle is inserted in the vicinity of the
tumor.
Percutaneous Microwave 4. The inner needle of the guide is removed.
Coagulation Therapy (PMCT) 5. The microwave electrode is inserted
through the outer needle of the guide into the
Methods (Fig. 17a.4) tumor.
6. The tumor is irradiated with microwaves
1. After the location of the tumor is con- at 80 W for 5 minutes. The microwave electrode
firmed, the puncture line from the point of is rotated axially during microwave irradiation
insertion of the electrode to the mass lesion is to prevent the coagulated tissue from adhering
to the tip of the electrode.
7. During microwave irradiation, the shaft of
the electrode and the guide needle are heated.
Therefore, to prevent the heat injury, the shaft
of the electrode and the guide needle are
cooled by cold saline using a cooling device.
8. The electrode and the outer needle of the
guide are removed. The puncture line is irradi-
ated with microwaves to prevent both bleeding
from the hepatic surface and cancer cells from
seeding along the puncture line when the elec-
trode is removed.
At each session, one to three electrode inser-
tions at different sites are performed for the
tumor. Microwave irradiation at 80W for 5
FIGURE 17a.3. Experimental study using resected minutes is performed for each electrode inser-
liver. With irradiation at 80 W for 5 minutes, the coag- tion. The end point is the hyperechoic changes
ulated area is elliptical, with maximal and minimal that cover the tumor and the neighboring non-
diameters of 3.5cm and 2.5cm, respectively. cancerous tissue to obtain a treated margin of
17a. Microwave Coagulation Therapy for Liver Tumors 221

FIGURE 17a.4. Method of percutaneous microwave coagulation therapy (PMCT). Black arrows show a
cooling device.

~5mm (Fig. 17a.5). When this change in the Microwave irradiation changes the low echo
echo texture is achieved, the initial session is image of a tumor to hyperechoic. However, it
completed. It is important that the approach is difficult to assess the therapeutic effect of
to the tumor should be multidirectional, for PMCT using US, because the echogenic change
example, subcostal and intercostal, to produce indicates not only tissue necrosis, but also water
extensive necrosis (Fig. 17a.6). vapor that is generated by the microwave irra-

FIGURE 17a.5. Sonographic changes before, during, was treated with two punctures and microwave
and after PMCT. The hepatocellular carcinoma irradiation. Note hyperechogenicity obscuring the
(HCC) measuring 3.0cm in diameter (white arrow) treated tumor.
222 T.Seki

FIGURE 17a.6. A 56-year-old man with an HCC that (dynamic CT, portal phase). PMCT induced a suffi-
was located in segment 8. CT obtained before and 1 cient treatment margin. The tumor and the sur-
week after PMCT treatment. Before treatment (CT- rounding area did not enhance. Accumulation of
A), the tumor site (white arrow, tumor size 1.8cm in iodized oil was observed in the tumor. The coagu-
greatest dimension) appears as an enhanced area. lated area was 3.7 cm in maximal diameter and
The tumor was treated with two subcostal and inter- 3.3 cm in minimal diameter.
costal probe insertions. One week after treatment

diation. Therefore, the therapeutic effect should organs also were excluded because of the pos-
be confirmed by dynamic computed tomogra- sibility of bile leakage or gastric or bowel per-
phy (CT), dynamic magnetic resonance imag- foration by heat injury.
ing (MRI), or enhanced US.
To assess the necrotic area, dynamic CT is
performed 2 to 3 days after each treatment Results of Percutaneous Microwave
in our hospital. When the initial session fails Coagulation Therapy
to obtain a desired treated margin, the next
One hundred thirty-one patients with hepatic
session is performed under US guidance with
cirrhosis and a solitary nodular HCC measur-
reference to postdynamic CT images taken
ing 3cm or less in diameter (tumor size ~2cm,
after the initial session. Thus, the total number
81 patients; >2 to ~3cm, 50 patients) who were
of sessions is determined by dynamic CT find-
admitted to Kansai Medical University and its
ings. In our experience, PMCT can be com-
affiliated hospital between September 1994 and
pleted with a single electrode insertion for most
March 2002 underwent PMCT alone.
HCCs measuring 2 cm or less in diameter
In patients with recurrence, we performed
(Fig. 17a.7). Likewise, three electrode insertions
transcatheter arterial chemoembolization
suffice for HCCs measuring more than 2 cm, but
(TACE) and PMCT, or TACE alone.
not more than 3.0cm in diameter.
Percutaneous microwave coagulation therapy
is performed twice weekly in our hospital when
indicated. In some patients, an iodized oil (4-
Survival
6mL, Lipiodol, Andre Guerbet, Aulnay-sous- The 5- and 7-year survival rates of patients with
Bois, France) is injected in the hepatic artery to HCC measuring 2cm or less in diameter
evaluate the grade of cancer. treated with PMCTwere 71 % and 56%, respec-
Tumors showing extrahepatic extension are tively. The 5-year survival rate of patients with
excluded from PMCT because of difficulty in HCCs measuring more than 2 cm, but no more
maintaining the puncture line. Similarly, tumors than 3.0cm in diameter, treated with PMCT
located near the gallbladder or gastrointestinal was 52%. At present, the 7-year survival rate of
17a. Microwave Coagulation Therapy for Liver Tumors 223

FIGURE 17a.7. A 72-year-old woman with HCC that tumor was treated with one needle pass. Sequence of
was located in segment 6 with underlying hepatic cir- enhanced CT (portal phase) scans (slice width: 1em,
rhosis (HCY). Before treatment (CT-A and CT-AP), A-D) one week after PMCT. The CT images show an
the tumor (white arrow, tumor size 2.0cm in greatest extensive avascular area surrounding the tumor.
dimension) enhanced with contrast material. The

patients with HCCs measuring more than 2 cm diameter after treatment with PMCT, the local
but no more than 3 cm in diameter with PMCT recurrence rate was 16%.
is unknown.
Combination Therapy
Cancer-Free Survival and Local (PMCT After TACE)
Recurrences
The 3- and 5-year cancer-free survival rates of Transcatheter arterial chemoembolization can
patients with HCC measuring 2cm or less in reduce the cooling effect for microwave thermal
diameter treated with PMCT were 40% and coagulation by decreasing hepatic arterial flow,
15%, respectively, while 3- and 5-year sur- and thus playa primary role in inducing tumor
vival rates of patients with HCCs measuring destruction. Furthermore, TACE with a combi-
more than 2.0cm but no more than 3.0cm in nation of iodized oil and gelatin sponge parti-
diameter with PMCT were 40% and 20%, cles can reduce the portal flow around the
respectively. tumor transiently by filling the peripheral portal
The incidence of definite local recurrence of veins around the tumor with injected iodized oil
HCC measuring 2.0cm or less in diameter after that passes through multiple arterioportal com-
PMCT within 2 years was 6%. On the other munications (6,7). This decrease of portal flow
hand, among patients with HCC measuring also may reduce the cooling effect. Moreover,
more than 2.0cm, but no more than 3.0cm in edematous changes (water retention) in the
224 T. Seki

tumor that include the surrounding area TABLE 17a.1. Complications associated with per-
induced by ischemia and inflammation after cutaneous microwave coagulation therapy.
TACE (8) can enlarge the coagulation area, Complication No. of cases
because microwaves act mainly on the watery Bleeding
component (water molecules) and produce Intraabdominal 2
dielectric heat (5). With an increasing watery Hemothorax 1
component, tissue necrosis induced by micro- Subcapsular 1
wave irradiation is enlarged (9). Furthermore, it Bile duct stricture 3
Hepatic infarction o
has been reported that PMCT with segmental Pleural effusion 4
hepatic blood flow (both arterial flow and portal Ascites 1
flow) occlusion produced extensive necrosis Tract seeding o
with a single needle insertion and subsequent 12/356 cases
microwave irradiation (10,11).

PMCT. Half of the patients felt some pain


Adverse Effects and during treatment, but it was not serious enough
Complications (Table 17a.l) to warrant treatment.
Local dissemination of the cancer cells along
At present, PMCT alone or in combination the tract was not encountered. However, com-
with TACE was performed in 356 patients plications such as intraabdominal bleeding,
having primary or recurrent nodules in our hos- hemothorax, subcapsular hematoma, intrahe-
pital. All patients complained of a slight heat patic duct stricture (Fig. 17a.8), pleural effusion,
sensation in the upper abdominal region during and ascites did occur.

FIGURE 17a.8. Intrahepatic bile duct stricture after (white arrow). About 3 months later, the stricture
PMCT. A 65-year-old man with an HCC that was was detected by the presence of dilated intrahepatic
located in segment 8. Single microwave irradiation bile ducts. The patient is being observed without
with 80W for 5 minutes was performed for the HCC treatment.
] 7a. Microwave Coagulation Therapy for Liver Tumors 225

There is a possibility that PMCT may induce It can be hypothesized that internal pressure
complications from direct heat injury by the within the HCC that is encased by a capsule is
microwave irradiation. Accordingly, close obser- elevated by the heat from RF ablation or
vation is required posttherapy. microwave coagulation. If this rise in the inter-
nal pressure is prolonged, the risk of tumor cells
escaping into the peritoneal cavity along the
The Choice Between PMCT and tract and/or hepatic vessels may be increased
Percutaneous Radiofrequency during ablation (17). Llovet et al (18) reported
that RF ablation with a cooled-tip probe for
Ablation (PRFA) Therapy for poorly differentiated HCCs located along the
HCC liver surface is associated with a high risk of
tract seeding. On the other hand, we have not
It is reported that lesions created by PRFA experienced tumor tract seeding associated
for HCC with one needle insertion for 10 to with PMCT. However, we believe that the short
20 minutes ranged in diameter from 3 to 4cm ablation time of PMCT may decrease the risk
(12-15). RFA requires a longer ablation time of tract seeding. Thus, PMCT using the newest
than microwave, but a larger area can be microwave coagulation system may be advan-
ablated per session. Recently, it has been tageous for small HCCs, although further clin-
reported that PRFA and PMCT have equiva- ical studies are required.
lent therapeutic effects, but that PRFA could be
achieved with fewer sessions. Thus, PRFA is
recommended for small HCCs (16). Currently Laparoscopic Microwave
PMCT is in early stages of development, and Coagulation Therapy (LMCT)
despite good coagulation capability, treated
areas induced by PMCT with one microwave When tumors have extrahepatic extension,
electrode insertion are small (1). However, we utilize laparoscopic microwave coagulation
PMCT can produce extensive necrosis with one therapy (LMCT). Recently, various microwave
needle insertion and 5 minutes microwave irra- electrodes and instruments for LMCT have
diation using the newer microwave coagulation been developed (Fig. 17a.9). LMCT is per-
systems. formed for HCCs that extend from the liver
Prolonged RF ablation may be stressful for edge (Fig. 17a.10). Thirty-six patients have been
patients and may result in tumor tract seeding. treated with LMCT in our hospital; five died of

FiGURE 17a.9. Microwave elec-


trodes (End-Angle electrode:
AZWELL, Osaka, Japan) for
laparoscopic microwave coagula-
tion therapy (LMCT). This elec-
trode has a flexible head for easy
needle insertion.
226 T. Seki

FIGURE 17a.l0. A 66-year-old man with an HCC that lesion. There were six needle insertions and micro-
was located in segment 3. Dynamic CT images wave irradiation (80W for 60 seconds) was per-
before (arterial phase) and after (portal phase) formed. After LMCT, the tumor and surrounding
LMCT. White arrow indicates the pretreatment area did not enhance.

the advanced HCC. However, at present, there 6. Kan Z, Sato M, Ivancev K, et al. Distribution and
are only two patients with definite local re- effect of iodized poppyseed oil in the liver after
currence. Using laparoscopic ultrasonography hepatic artery embolization: experimental study
(Laparo-US) can be quite difficult, and skill and in several animal species. Radiology 1993;186:
training are required (19). In cases of HCCs 861-866.
7. Chung JW. Transcatheter arterial chemoem-
that exist under the diaphragm and cannot be
bolization of hepatocellular carcinoma. Hepato-
visualized under laparoscopy and Laparo-US, Gastroenterology 1998;45(suppl 3):1236-1241.
thoracoscopy is used (20). 8. Yoshioka H, Nakagawa K, Shindou H, et al. MR
imaging of the liver before and after trans-
References catheter hepatic chemo-embolization for hepa-
tocellular carcinoma. Acta RadioI1990;31:63--67.
1. Seki T, Wakabayashi M, Nakagawa T, et al. 9. Seki T, Tarnai T, Nakagawa T, et al. Combination
Ultrasonically guided percutaneous microwave therapy with transcatheter arterial chemoemboli-
coagulation therapy for small hepatocellular zation and percutaneous microwave coagulation
carcinoma. Cancer 1994;74:814-825. therapy for hepatocellular carcinoma. Cancer
2. Seki T, Wakabayashi M, Nakagawa T, et al. 2000;89:1245-1251.
Percutaneous microwave coagulation therapy 10. Murakami T, Shibata T, Ishida T, et al. Percuta-
for small hepatocellular carcinoma: Comparison neous microwave hepatic tumor coagulation
with percutaneous ethanol injection therapy. with segmental hepatic blood flow occlusion in
Cancer 1999;85:1694-1702. seven patients. AJR 1999;172:637-640.
3. Tabuse K, Katsumi M. Application of microwave 11. Shibata T, Murakami T, Ogata N. Percutaneous
tissue coagulator to hepatic surgery. The hemo- microwave coagulation therapy for patients with
static effects of spontaneous rupture of primary and metastatic hepatic tumors during
hepatoma and tumor necrosis. Arch Jpn Chir interruption of hepatic blood flow. Cancer 2000;
1981;50:571-579. 88:302-311.
4. Tabuse K, Katsumi M, Kobayashi Y, et al. 12. Patterson EJ, Scudamore CH, Owen DA, Nagy
Microwave surgery: hepatectomy using micro- AG, Buczkowski AK. Radiofrequency ablation
wave tissue coagulator. World J Surg 1985;9: of porcine liver in vivo. Effects of blood flow and
136-143. treatment time on lesion size. Ann Surg 1998;
5. Tabuse K. Basic knowledge of a microwave 227:559-565.
tissue coagulator and its clinical applications. 13. Goldberg SN, Hahn PF, Tanabe KK, et al. Per-
J Hepatol Bi! Pancr Surg 1998;5:165-172. cutaneous radiofrequency tissue ablation: Does
17a. Microwave Coagulation Therapy for Liver Tumors 227

perfusion-mediated tissue cooling limit coagula- arterial chemoembolization and percutaneous


tion necrosis? J Vasc Intervent Radiol 1998;9: radiofrequency ablation in the primary tumor
101-111. region. Eur J Gastroenterol Hepatol 2001;13:
14. de Baere T, Denys A, Wood BJ, et al. Radiofre- 291-294.
quency liver ablation: experimental compara- 18. Llovet JM, Vilana R, Bru C, et al. Increased risk
tive study of water-cooled versus expandable of tumor seeding after percutaneous radiofre-
systems. AJR 2001;176:187-192. quency ablation for single hepatocellular carci-
15. Goldberg SN, Gazelle GS. Radiofrequency tissue noma. Hepatology 2001;33:1124-1129.
ablation: physical principles and techniques 19. Ido K, Isoda N, Kawamoto C, et al. Laparoscopic
for increasing coagulation necrosis. Hepato- microwave coagulation therapy for solitary
Gastroenterology 2001;48:359-367. hepatocellular carcinoma performed under
16. Shibata T, Iimuro Y, Yamamoto Y, et al. Small laparoscopic ultrasonography. Gastrointest
hepatocellular carcinoma: comparison of radio- Endosc 1997;45:415-420.
frequency ablation and percutaneous microwave 20. Yamashita Y, Sakai T, Maekawa T, Watanabe K,
coagulation therapy. Radiology 2002;223:331- Iwasaki A, Shirakusa T. Thoracoscopic transdi-
337. aphragmatic microwave coagulation therapy for
17. Seki T, Tarnai T, Ikeda K, et al. Rapid progression a liver tumor. Surg Endosc 1998;12:1254-1258.
of hepatocellular carcinoma after transcatheter
17b
Microwave Ablation:
Surgical Perspective
Andrew D. Strickland, Fateh Ahmed, and David M. Lloyd

The Field Effect of able applicator or probe as a "field" around its


tip. Direct heating of water molecules occurs
Microwave Heating within the whole microwave field, not simply by
conduction of heat from the surface of a hot
Thermal destruction by microwaves has been probe. A whole spherical area, perhaps the size
used effectively for many years for ablation of of a tennis ball, is heated simultaneously and
both small liver metastases and primary lesions. uniformly within minutes. Heating is not reliant
Microwaves produce effective ablation without on conduction through the tissues. Cytotoxic
islands of viable cells in a rapid and repro- increases in temperature are reached rapidly.
ducible fashion. The microwave region of the Beyond the microwave field, however, heating
electromagnetic spectrum is well suited to such of the adjacent tissue does occur by thermal
a role due to the efficient conversion of elec- conduction.
tromagnetic energy to heat. This translation The distinction between conductive heating
of energy is a result of the strong interaction and direct field heating is important, as con-
between polar molecules and microwaves duction through water-laden tissue is slow and
that causes oscillation of molecules, which is inefficient, particularly when the effects of local
expressed as heat. blood flow act as a heat sink. The interaction
Water is a highly polar molecule, abundant in between the microwave field and water mole-
both normal liver tissue and hepatic neoplasms; cules of the target tissue within that field occurs
it is the interaction between the microwaves very rapidly; consequently, the temperature
and water that is principally responsible for the climbs quickly. Theoretically, microwave coagu-
rise in temperature. The interaction between lation should be the most efficient method for
water and the entire range of frequencies in the the destruction of large liver tumors.
microwave bandwidth is particularly strong. It Until recently the production of large, pre-
is no coincidence that the fastest form of dictable ablations with microwave devices has
heating foodstuffs in the domestic kitchen is the not been possible unless multiple insertions
microwave oven, principally due to the interac- of a probe or needle were used, similar to
tion between water molecules and microwaves. radiofrequency ablation (RFA). This constraint
Another advantage of microwave ablation is has limited its use among liver surgeons and
the manner in which the heating occurs. Unlike interventional radiologists for ablation of col-
the alternative ablative devices such as radio- orectal metastases and small primary hepato-
frequency or cryotherapy, the passage of heat is cellular carcinomas in cirrhotic patients.
not solely reliant on conduction. The funda- Currently, the treatment of larger tumors has
mental reason why microwave energy is unique been possible onl~ qy multiple placements of
and efficient is that it is transmitted from a suit- a microwave probe that produce overlapping

228
17b. Microwave Ablation: Surgical Perspective 229

volumes of ablation. This is undesirable, as extremely efficient heating of tissue within its
the technique requires highly skilled, accurate microwave field.
placements of the probe to avoid leaving The data gained from the experiments into
islands of viable islands of cells in the target the microwave properties of tumor tissue and
tumor. It also means that many patients will normal liver parenchyma have given physicists
undergo multiple treatment sessions. more knowledge of the electromagnetic prop-
erties of target tissues and how they change
during treatment, Using this information, accu-
The Development of aNew rate computer models have been constructed to
Microwave Applicator for Large test the efficacy of novel applicator designs,
thereby avoiding the need to "bench-test"
Liver Tumor Ablation every new probe. The models assess radiation
of the microwave into the tissues and the
Over the last decade a new form of microwave degree of reflection that will occur.
treatment applicator, the microwave endome-
trial ablation (MEA) applicator, has been
developed by Microsulis Medical Ltd., UK in Animal Experiments on
conjunction with scientists at the University of
Bath, UK. The initial focus was in the field Production of Large Ablation
of gynecology. A microwave applicator was Volumes by Microwave Energy
designed specifically to treat women with men-
orrhagia using an 8.5-mm-diameter antenna Female pigs (body weight 45-50 kg) were anes-
that was able to produce a heated microwave thetized and underwent a laparotomy to allow
field from a generator delivering a frequency good exposure of the liver. Once adequately
of 9.2 GHz. Microwave endometrial ablation exposed, the novel microwave applicators were
now is an established method to destroy the inserted into the hepatic parenchyma and abla-
endometrium. It has been successfully used to tions were carried out at varying times (1 to 3
treat more than 30,000 patients worldwide and minutes) and power settings (45 to 250W). The
the safety and efficacy of this treatment has most remarkable finding was that the
been extensively validated in randomized con- microwave equipment was able to produce
trolled trials (Fig. 17b.1) (1). The uniqueness large volume ablations (5 to 8cm in diameter)
of the applicator is its ability to transmit quickly, and were spherical in nature. The
microwave energy via a coaxial cable with results were reproducible, and there was a
linear relationship between the size of burn and
the power output for any given time (2,3). Thus,
accurate treatment volumes could be calculated
for any given tumor size. The animals tolerated
the production of multiple liver ablations
extremely well. A large, 7-cm liver ablation with
a single insertion is seen in Figure 17b.2, pro-
duced by using the 6.4-mm probe, powered at
150W, at a frequency of 2.45 GHz.

Clinical Studies with Large


Ablations
FIGURE 17b.1. The microwave endometrial ablation Although microwave liver ablation has been
(MEA) applicator designed by Microsulis Medical used extensively in the Far East over the past
Ltd., UK. 20 years to treat primary liver tumors in cir-
230 A.D. Strickland et al

A B
FIGURE 17b.2. Experimental liver ablation in a large holder (left) with the production of a 7-cm-
animal model. The applicator is seen (with centime- diameter ablation within 3 minutes (B).
ter markings) (A) adjacent to thermocouples in a

rhotic patients, the results have been mixed, that can potentiate the "heat-sink" effect, as the
and limited by the size of the needle applicator high energy has to travel through tissues (bile
that can treat only small lesions «2 cm) unless ducts and vessels) to complete the electric
multiple punctures are utilized. Developments circuit.
in radiofrequency (RF) probes are encourag- To date, our experience has included 22
ing, but RF also is limited because large tumor patients with inoperable liver cancer who were
ablation (>5 cm) is associated with high recur- entered into a clinical study. The study was
rence rates. approved by our institution's ethical commit-
We have used the large microwave applica- tee. Six patients had inoperable primary liver
tor clinically with encouraging early results. cancer, 15 had inoperable colorectal metas-
There is no need for a conduction plate to be tases, and one had multiple neuroendocrine
placed on the body, thereby eliminating the tumors. The age range was 38 to 79 years with
potential for skin complications that can occur a median age of 72 years. Four patients under-
with RFA. It also reduces the chance of bile went simultaneous resection of the colorectal
duct damage caused by RF electric currents primary, and 12 underwent liver resection as
well as microwave ablation.
Patients with tumors greater than 5 cm in
diameter were treated for 3 or 4 minutes using
the lower frequency of 2.45 GHz with the
6.4-mm-diameter probe at 150W. Those with
smaller tumors were treated for between 1 and
3 minutes at 9.2GHz frequency at 45W (Fig.
17b.3). Over 60 total ablations were performed,
with the mean number of treatments being four
per patient (range 1-9).
No complications were seen, and no early
recurrences have occurred at the ablation sites
with a mean follow-up of 12 months. There
were two deaths in the colorectal group and
one in the primary hepatocellular carcinoma
(HCe) group, which occurred within 30 days.
One patient treated for a large, 5.5-cm HCC
was readmitted to the hospital with liver
FIGURE 17b.3. Microwave ablation of a liver tumor decompensation 5 weeks posttreatment. None
using the novel microwave applicator. A 2-cm tumor of these deaths was directly attributable to the
is seen surrounded by a large area of ablation. microwave ablation. The two patients who died
17b. Microwave Ablation: Surgical Perspective 231

in the colorectal group were elderly and frail years. His a-fetoprotein levels fell from 6000
and were awaiting triple coronary bypass graft- units preoperatively to 40 units within a few
ing for severe ischemic heart disease. Both of months after the ablation. He had a slow-
these patients were rejected for cardiac surgery growing pleural metastasis that probably
because of the presence of inoperable liver accounted for the slightly raised a-fetoprotein.
tumors. The microwave ablations in both of Follow-up computed tomography (CT) scans
these patients were minimal, as only a few 2-cm confirmed he had complete regression of his
metastatic lesions were treated, and did not liver primary.
contribute to the deaths of these patients. A second patient was deemed to have inop-
The mean survival for the 22 patients in this erable hepatic metastases from a colorectal
initial study group was 12 months, with most carcinoma. This 59-year-old woman failed to
patients dying of causes unrelated to their liver respond to chemotherapy. There were five large
treatment. Several patients died of sepsis sec- tumor deposits in the right liver and one large
ondary to urinary tract infection or pneumonia one (6cm diameter) within the left lateral
8 or 9 months after discharge from hospital, and segment (Fig. 17b.5).
other patients developed thoracic metastases. The patient underwent resection of four seg-
There was no evidence that any of these ments (segments 2, 3, 5, and 6) with four metas-
patients developed local recurrence at the site tases. Microwave ablation of the two lesions in
of a microwave ablation, although some segments 7 and 8 was performed. She did well
patients developed liver tumors in other parts following the surgery, and remained in the hos-
of the liver. pital for 14 days. The follow-up scans taken at
An example of the microwave procedure is 2 months postablation showed no viable recur-
demonstrated in a 74-year-old retired general rence in the ablation areas or the remaining
practitioner with known liver cirrhosis who liver (Fig. 17b.6).
developed a 6.5-cm primary liver tumor in the The new applicator can be used for both
right lobe of the liver (Fig. 17b.4). laparotomy and laparoscopy. Large ablations
Although a trial dissection was attempted, it can be treated. For example, a 37-year-old
was abandoned because of bleeding secondary woman presented with pain in her right upper
to portal hypertension. During the procedure, quadrant, and was found to have a 6.4-cm
it was elected to treat the tumor with the 6.4- benign adenoma in the middle of the right lobe.
mm probe with a frequency of 2.45 GHz for 3 Rather than subject her to a formal right hemi-
minutes. No bleeding occurred at the time of hepatectomy, she was treated laparoscopically
ablation. The patient has remained well for 2% using a 5.5-mm microwave probe. She under-

A 8
FIGURE 17bA. (A) Coronal magnetic resonance (arrows). (B) Two years after microwave ablation
imaging (MRI) scan of a patient with a 6.5-cm treatment, the lesion (arrow) remains small and inac-
primary liver tumor (HCC) in the dome of the right tive on the contrast-enhanced computed tomogra-
hepatic lobe is seen as the lower-intensity mass phy (CT) scan.
232 A.D. Strickland et al

A B
FIGURE 17b.5. (A,B) MRI scan of a patient with six large colorectal metastases in the liver. Three tumors in
segments 5,6, and 8 are shown. The 6-cm tumor in segments 2 and 3 is not shown.

A B
FIGURE 17b.6. (A,B) Follow-up MRI scans show two areas of ablated lesions (segments 7 and 8) in the hyper-
trophied liver.

A B
FIGURE 17b.7. (AB) The preablation MRI scan of a 5.5-cm adenoma is seen on the left as a high-
intensity mass (arrow). Six months following microwave ablation, a scar (arrow) remains.
17b. Microwave Ablation: Surgical Perspective 233

went ablation using 150W at 2.45GHz for only also feasible laparoscopically. Thinner probes
2 minutes. The preoperative and 6-month for percutaneous treatments are being devel-
follow-up MRI scans of her liver are shown in oped as well.
Figure 17b.7.
References
Conclusion 1. Cooper KC, Bain C, Parkin DE. Comparison of
microwave endometrial ablation and transcervi-
These early data confirm that microwave abla- cal resection of the endometrium for treatment of
tion is safe, very quick, and efficient to destroy heavy menstrual loss: a randomised trial. Lancet
1999;354:(9193):1859-1863.
liver tumors within minutes. Large ablations
2. Swift B, Strickland A, West K, Clegg P, Cronin N,
can be achieved quickly and safely. We have not
Lloyd DM. The histological features of microwave
experienced any specific microwave-related coagulation therapy: an assessment of a new
complications during or after surgery. Large- applicator design. Int J Exp Pathol 2003;84(1):
volume ablations greater than 5cm in diameter 17-30.
are achieved, and ablations of 8 cm in diameter 3. Strickland AD, Clegg PJ, Cronin NJ, et al. Exper-
may be accomplished. While large ablations imental study of large volume microwave ablation
have been achieved at open surgery, they are in the liver. Br J Surg 2002;89(8):1003-1007.
18
Percutaneous Laser Therapy of Primary
and Secondary Liver Tumors and Soft
Tissue Lesions: Technical Concepts,
Limitations, Results, and Indications
Thomas 1. Vogl, Ralf Straub, Katrin Eichler, Stefan Zangos, and Martin Mack

Percutaneous laser therapy is one of the point of view, therapeutic decisions have to be
general percutaneous ablation therapy strate- made according to curative, palliative, and
gies. In principle, laser therapy is possible using symptomatic indications.
fibers or by laser-induced thermotherapy This chapter presents the technical require-
(UTT). The UTI procedure currently is ments, the methodology, and the results of
performed at laparotomy, by laparoscopic MRI-guided UTI in the field of liver and head
control intraoperatively, or percutaneously. and neck tumors, and general oncologic issues.
Monitoring is essential for percutaneous inter- The liver as a large solid organ is very suit-
ventions; magnetic resonance imaging (MRI) able for interventional radiology procedures,
has proven to be the most reliable thermal due to the short transcostal access and its excel-
measure, while ultrasound has been unable to lent functional capacity. It is the most common
show an adequate estimation of the thermal site of metastatic tumor deposits, especially for
changes induced by the applied energy. Laser- colorectal cancer, which is the third leading
induced thermotherapy makes available a pho- cause of death in Western communities, out-
tothermal tumor destruction technique that numbered only by lung and breast cancer. At
allows solid tumors within parenchymal organs the time of death, approximately two thirds of
to be destroyed. The laser energy is transmitted patients with colorectal cancer present with
via thin optic fibers and causes a well-defined liver metastases.
area of coagulative necrosis. This effect results As metastatic disease is so prevalent in the
in destruction of tissue by direct heating, while liver, new treatment protocols currently are
limiting damage to surrounding structures. under investigation. Laser-induced thermother-
Magnetic resonance imaging has proven to be apy recently has been applied as a minimally
an ideal clinical instrument to define the exact invasive technique to treat liver metastases. The
position of the optical fibers in the target area, percutaneous approach avoids laparotomy and
provides real-time monitoring of the thermal uses local anesthesia with outpatient manage-
effects, and the subsequent evaluation of the ment, which are the main advantages of this
extent of coagulative necrosis. new therapy.
Several clinical indications for the use of min- After cure of the primary tumor, hepatic
imally invasive laser applications currently are metastases have a decisive influence on sur-
under investigation. By far the most frequent vival. Therapeutic strategies for malignant liver
use is in the liver, followed by soft tissue tumor lesions are based on a number of factors,
ablation, ablation and of lung, breast, head and such as the underlying primary tumor, location,
neck, and renal tumors. From the oncologic stage of the tumor, and general factors, such as

234
18. Percutaneous Laser Therapy 235

age and any existing concomitant illness. The range of the resulting temperature rise is
For example, when hepatocellular carcinoma not restricted to the optical penetration depth,
(HCC) is curable, liver resection, hemihepatic but is substantially extended by thermal con-
resection, or liver transplant is the treatment of duction. Normal cells are less sensitive to
choice. If there are contraindications, transarte- thermal exposure, while malignant cells are
rial chemoembolization combined with local more sensitive. The altered metabolic state of
alcohol injection is used as a palliative thera- malignant cells with pronounced hypoxia
peutic strategy. Interstitial procedures such as causes this high sensitivity.
LITT show a high rate of controlling the tumor, To do justice to the coagulation of three-
and are being evaluated clinically. dimensional tumor geometry, it must be possi-
Strategies for liver metastases are consider- ble to heat an approximately spherical volume
ably more complex. Up to now, surgical resec- of tissue throughout. For this reason, applica-
tion of solitary lesions has been the only tion systems of defined space radiation have
potential curative treatment. However, the high been developed, the distal ends of which are
rate of intrahepatic relapse, and the possible designed such that the result is an even cir-
potentiation of intrahepatic growth of metas- cumference of radiation. With conventional
tases because of tumor stimulation caused by applicators, almost spherical coagulation zones
release of growth-stimulating substances, are with a diameter of 20 to 25 mm are achieved.
considered problematic. For these reasons, over Apart from the applicator geometry, the radia-
the past few years there has been great interest tion capacity and time are crucial parameters
in development of interstitial procedures such for the dimensions of the coagulation zone, as
as laser coagulation or radiofrequency ablation. are specific tissue properties such as optical
parameters and perfusion rate.
The temperature-dependent effects of the
Laser Application Techniques laser light on the tissue include enzyme induc-
tion, edema formation, and membrane relax-
For the theoretical and clinical use of UTT, the ation in a temperature range of 40° to 45°C.
tissue-destroying effect is dependent on the At 60°C protein denaturation takes place, at
choice of both radiation capacity and time. This 80°C collagen denaturation, and over 150°C
means that parameters must be preselected in carbonization.
such as way that all tumor cells, if possible, are
exposed to the coagulative effect. There also
must be a safety margin of at least 10 mm in
width around the tumor. Adjacent sensitive
Applicators and Application
structures must not be damaged. Selecting the
Techniques
right laser is based on an optimal depth effect Tissue carbonization must be avoided to
in tissue that is determined by the absorption achieve large-volume coagulation zones and to
properties of water and hemoglobin. This be able to guarantee safe application. The crit-
can be achieved with a wavelength between ical value is a power density of approximately
1060 and 1200nm. The neodymium: yttrium- 5W/cm2 • Depending on the applicator, this
aluminum-garnet (Nd:YAG) and semiconduc- means laser power of 3 to 10 W. At this point,
tor lasers with wavelengths of 1060 nm or 900 migration of the photons deep into the tissue
nm fulfill these requirements. The Nd:YAG through absorption into the resulting "carbon
laser is the solid-state laser used most often, layer" is impeded, and only "a hot spot" is pro-
and it comes with a wavelength of 1064nm. duced. The heat diffusion that is still active
The laser can be both pulsed and operated con- limits the UTI zone to a small volume (diam-
tinuously, and supplies output power of up to eter of 1 cm).
WOw. The tissue-dependent penetration depth To avoid the carbonization effect that greatly
of the photons is an essential parameter of the reduces the volume, a dome-shaped applicator
absorption in various depths of tissue layers. (scattering dome) has been developed. The
236 T.J. VagI et al

laser- FIGURE 18.1. Illustration of the


silicafiber applicator internally cooled power laser
(400 J.lm) (active zone)
system.

double-tube
cooling sheath protective catheter
(inflow)
cooling
(outflow)

scattering dome applicator can achieve the A method of modifying the size and mor-
maximum penetration depth of the photons phology of necrosis is a multiapplication with
into the tissue. Because of absorption and unifocal access ("pull-back" technique). After
convection, additional heat propagation results the end of the first heat application, the laser
in deeper heating and hence "expansion" of fibers are withdrawn by 1 and 2 em through the
the applicator, through which the diffusion percutaneous access point in the puncture
processes of the heat propagation becomes pathway, while further heat application is
dominant. It is technically easy and effective to applied.
adapt the scatter applicator to the therapeutic As well as monoapplicators, multiapplicators
situation (Fig. 18.1). Radiation times over 30 can be utilized (multiapplications with multi-
minutes do not lead to any substantial addi- focal access). Here, two or even up to four
tional expansion of the damaged tissue (diffu- applicators are laid parallel and operated
sion equilibrium). simultaneously. Prerequisites are the corre-
An essential step in clinical UTT is the sponding number of laser devices or beam split-
development of application systems that can be ters that help treat larger malignant lesions
applied percutaneously, laparoscopically, or more rapidly. The disadvantage is the higher
operatively. First, a conventional system was number of punctures that are required.
available that consisted of a scattering dome
applicator, a sluicing system, and a ther-
mostable sheathed catheter, via which the UTI
applicator was positioned (Fig. 18.2A).
Treatment Monitoring
Development of an irrigation system further Magnetic resonance tomography (MRT) and
expanded laser-induced necrosis, and optimized MR thermometry (MRTE) are the optimum
the treatment. This system has been developed imaging methods to monitor treatment. Multi-
from a 9-French sluicing system with centime- planar representation and the high soft tissue
ter markings, and a 7-French sheathed catheter contrast of MRT highlight the value of these
with double lumina for irrigation. A sterile salt techniques (Fig. 18.2).
solution at room temperature is a proven irri- Magnetic resonance sequences that empha-
gation medium. With a pump system integrated size perfusion and diffusion are temperature
into the laser, irrigation rates of 30 to 60mL per dependent and thus can be used for noninva-
minute can be reached, thus providing reliable sive temperature measuring. These parameters
cooling of the applicator zone. include the diffusion coefficient of water, the
The following application techniques permit photon resonance frequency or chemical shift,
further treatment optimization; for the mono- and the Tl relaxation time. Due to the rela-
application, an application system is placed in tively low sensitivity with regard to motion arti-
the lesion percutaneously and is removed after facts, their wide availability, and the speed of
application of heat. data acquisition, thermosensitive Tl-weighted
~
"d
(1l
;::l
S-
III
::I
(1l
o
~
h;'
(1l
...'"
...~

'<

A-C

D-G
FIGURE 18.2. (A) A 35-year-old woman with liver metastasis from a col- heated area. (E) T1-weighted thermosensitive gradient echo sequence in
orectal carcinoma (initial tumor stage pT3, N1, M1). The T1-weighted ther- sagittal tomographic orientation in the 16th minute. Wider reduction in signal
mosensitive gradient echo sequence in axial tomographic orientation shows intensity in the heated area. (F) T1-weighted thermosensitive gradient echo
the metastasis in liver segment 8 before starting the laser. (B) T1-weighted sequence in sagittal tomographic orientation in the 20th minute. Low signal
thermosensitive gradient echo sequence in axial tomographic orientation in intensity in the heated area. (G) T1-weighted spin echo sequence in axial
the 1st minute. Clear drop in the signal intensity in the heated area. (C) T1- tomographic orientation after the administration of gadolinium-diethylene-
weighted thermosensitive gradient echo sequence in sagittal tomographic triamine pentaacetic acid (DTPA) (O.lmL per kg body weight). Sharp
orientation in the 3rd minute. Further drop in signal intensity in the heated demarcation of the laser-induced thermotherapy (LITT)-induced necrosis
area. (D) T1-weighted thermosensitive gradient echo sequence in sagittal with a safety margin surrounding the lesion.
N
...,
tomographic orientation in the 7th minute. Drop in signal intensity in the
-..l
238 T.J. Vogi et al

MRTE sequences are used for clinical imple- optimize T1-weighted sequences (Figs. 18.3 and
mentation of hepatic LITT. The longitudinal or 18.4).
spin-lattice relaxation time of a tissue is tem- Computer-aided treatment planning that has
perature dependent, and a local rise in temper- been developed through in vivo comparative
ature results in a signal drop in the MRT image. tests is available to further aid therapy. This
In vivo examinations and the findings of ours makes it possible to calculate the optimum
and other teams have shown a virtually linear parameters for radiation capacity and time for
correlation between the drop in signal in the each individual fiber before carrying out the
image and the temperature. Appropriately laser treatment. The progress of the current
weighted gradient echo sequences [fast low- temperature and the irreversible damage dis-
angle shot (FLASH) and turbo-FLASH] with tribution can be monitored simultaneously as
measuring times between 6 and 15 seconds in progress of treatment with real-time simula-
breath-hold technique have proved suitable tion, so that magnetic resonance tomography
for depicting the laser-induced temperature with indirect monitoring is available as an
changes ranging from 60 to 1l0°e.
0
extension of virtual on-line monitoring.
Before and after LITT a test of T1- and Implementing and evaluating computer-
TI-weighted spin echo and gradient echo aided thermoplanning for LITI applications is
sequences is used for treatment planning and costly. The expected irreversible damage zone
control. In addition, contrast agents [0.1 mmol depends on various complex parameters.
gadolinium-diethylenetriamine pentaacetic Influencing factors are laser capacity, radia-
acid (Gd-DTPA) per kilogram of body weight] tion time, applicator characteristics, and optical

FIGURE 18.3. UTI of pleomorphic adenoma. (A) changes. Right image: The contrast-enhanced image
Thermo-turbo FLASH image (TRffEffI/flip angle 1 week after UTI shows decreasing postablation
= 7/3/400/8°) demonstrates the tumor recurrence reactive changes and an increase in size of zone of
(arrows) of the pleomorphic adenoma in the pre- coagulative necrosis (N). (D) Left image 4 weeks
styloid compartment of the right parapharyngeal after UTT, middle image 3 months after UTI, right
space nearly isointense to muscle tissue. A subzygo- image 2 years after UTI. Left image: Contrast-
matic approach was used for positioning of the laser enhanced (0.1 mmol/kg h w. Gd-DTPA) Tl-weighted
applicator. Note the signal loss of the laser applica- SE image (TRffE = 700/15) demonstrates a further
tor due to the magnetite marker. The active zone decrease of the reactive changes in the border of the
(length 2cm) of the laser applicator starts 1cm treated recurrent tumor (arrows). However, in the
before the end of the magnetite marker. (B) Thermo- anterior portion of the tumor pathologic contrast
turbo FLASH image obtained 12 minutes after start- enhancement still could be visualized. Therefore, a
ing the laser shows a signal loss around the active second UTT treatment was performed to treat this
zone of the laser applicator (arrow) after an increase part of the recurrent tumor. Middle image: The con-
of tissue temperature due to an increase in T1 relax- trast-enhanced image 3 months after LITT demon-
ation time. (C) Left image before UTT, middle strates the induced coagulative necrosis (N) with
image 2 days after UTT, right image 1 week some residual reactive changes in the border of the
after UTT. Left image: The contrast-enhanced lesion. Right image: The contrast-enhanced image 2
(O.lmmol/kgb.w. Gd-DTPA) Tl-weighted spin echo years after UTI shows decreasing postablation
(SE) image (TRffE = 700/15) demonstrates the reactive changes (arrows). (E) The plain Tl-
recurrent pleomorphic adenoma in the pre-styloid weighted SE image (TR/TE = 700/15) obtained 4
compartment of the left parapharyngeal space with years after laser treatment demonstrates mainly
a strong contrast enhancement, displacing the residual scar tissue in the pre-styloid compartment
internal carotid artery on the left side posteriorly of the left parapharyngeal space. (F) The contrast-
(arrows). Middle image: Contrast-enhanced image 2 enhanced (0.1 mmol/kg hw. Gd-DTPA) Tl-weighted
days after UTI with 5.8W over 20 minutes demon- SE image (TRffE = 700/15) demonstrates no patho-
strates the induced coagulative necrosis (N) with logic contrast enhancement in the pre-styloid com-
strong enhancement in the border of the lesion partment of the left parapharyngeal space, indicating
most likely representing postinterventional reactive lack of viable tumor.
240 T.J. Vogl et al

and thermal tissue parameters such as tissue Imaging During Therapy


perfusion and blood flow. Initial applications
of this type of system lead us to expect After informing the patient about the advan-
further improvements in precision during tages, disadvantages, and potential compli-
treatments. cations of UTI, consent is obtained. The
metastasis is localized on computed tomo-
graphic scans and the injection site is infiltrated
Standard Laser-Induced with 20mL of 1 % lidocaine. The laser appli-
cation system is inserted under computed
Thermotherapy Procedure to tomography (CT) guidance using the Seldinger
Treat Malignant Liver Tumors technique. The patient then is transferred to
MRI. After the patient is positioned on the
Laser coagulation uses an Nd:YAG laser light MRI table, the laser catheter is inserted into the
with a wavelength of 1064nm (MediLas 5060, protective catheter. The MR sequences are
MediLas 5100, Dornier Germering, Germany). performed in three perpendicular orientations
It is delivered through optic fibers whose tips before and during UTI; MRI is obtained every
end in a specially developed diffusor. Originally, 30 seconds to assess the progress in heating of
a diffusor tip with a glass dome of 0.9-mm diam- the lesion and the surrounding tissue. Heating
eter that was mounted at the end of a 10-m-Iong is manifested as signal loss on the T1-weighted
silica fiber (diameter 400llm) was used. Since gradient-echo images as a result of the heat-
2000, a flexible diffuser tip has been used with a induced increase of the T1 relaxation time.
diameter of 1.0 mm; this facilitates laser applica- Depending on the geometry and intensity of
tions since the risk of damage to the diffuser tip the signal loss and the speed of heat distribu-
has decreased to almost zero. The active length tion, readjustment can be made in the position
of the diffuser tip ranges from 20 to 40mm in of the laser fibers, the laser power, and the
length. The laser power is adjusted to 12W per cooling rate. The treatment is stopped after total
centimeter active length of the laser applicator. coagulation of the lesion, and a safety margin
The laser application kit (SOMATEX, from 5 to 15 mm surrounding the lesion is visu-
Berlin, Germany) consists of a cannulation alized on MR images. After switching off the
needle, a sheath system, and a protective laser, T1-weighted contrast-enhanced FLASH-
catheter that prevents direct contact of the laser two-dimensional (2D) images are obtained to
applicator with the treated tissues and allows verify the necrosis. After the procedure, the
cooling of the tip of the laser applicator. The puncture channel is sealed with fibrin glue.
closed end of the protective catheter enables Follow-up examinations using plain and con-
complete removal of the applicator even in the trast-enhanced MRI sequences are performed
unlikely event of damage to the fiber during at 24 to 48 hours, and every 3 months following
treatment. This simplifies the procedure and the UTI procedure. Quantitative and qualita-
makes it safer for the patient. tive parameters, including size, morphology,
The laser itself is installed outside the MR signal behavior, and contrast enhancement are
examination room, and the light is transmitted evaluated to decide whether treatment can be
through a 10-m-Iong optic fiber. All patients are considered successful or whether subsequent
examined using an MRI protocol including gra- treatment sessions are required.
dient-echo (GE) Tl-weighted plain and con-
trast-enhanced scans (Gd-DTPA O.lmmollkg
body weight). T1- and T2-weighted sequences
Qualitative and Quantitative
are obtained to localize the target lesion and
Evaluation
plan the interventional procedure. The scan-
ners are a conventional 1.5-T magnet system Laser-induced effects are evaluated by compar-
(Siemens, Erlangen, Germany) and a 0.5-T ing images of lesions and surrounding liver
system (Elscint Privileg). parenchyma obtained before and after treat-
18. Percutaneous Laser Therapy 241

c
A

B o

FIGURE 18.4. Laser treatment of a parastomal recur- orientation verifies the position of the application
rent squamous cell carcinoma after laryngectomy. set. A specially designed magnetite marker is
(A) Contrast-enhanced Tl-weighted SE image already inserted into the application set (arrows).
demonstrates recurrent tumor on the right side with (D) Thermo-FLASH-2D image 4 minutes after start-
an infiltration of more than 180 degrees of the ing the laser demonstrates the close relationship of
circumference of adjacent vascular structures with the induced necrosis to the vascular structures and
strong contrast enhancement (arrows). (B) The CT the esophagus (circle). (E) Contrast-enhanced Tl-
image demonstrates the positioning of the laser weighted SE image obtained 6 months after LITT
application set with the sheath system (arrows) and treatment shows an excellent result of this combined
the protective catheter. A power application set was treatment protocol.
used. (C) The FLASH-2D MR image in axial slice
242 T.J. Vogi et ai

ment, and at follow-up examinations. Tumor LITT for Soft Tissue Tumors
volume and volume of coagulative necrosis are
calculated using three-dimensional MR images • Recurrent head and neck tumors
and measurements of the maximum diameter • Tumor recurrence in the pelvis
in three planes (A, B, and C). The volume is cal- • Lymph node metastases in the abdomen,
culated using the formula (A x B x C) x 0.5. the head and neck region, and the
The results are tested for significance using the retroperitoneum
analysis of variance (ANOVA) test. Survival Exclusion Criteria for LITT
rates are calculated using the Kaplan-Meier
method. • Extensive extrahepatic tumor spread
• Contraindications for MRI (pacemaker)
• Ascites, apparent infection, coagulation
disorder
Indications • Diffuse and multiple metastases
The primary therapeutic goal is defined as local
tumor control in patients with limited hepatic Comparison with Alternative
malignant tumor. The majority of patients in Methods
our experience have had liver metastases from
colorectal cancer. However, solitary hepatic Surgery
metastases from other primary tumors, like
breast cancer, carcinoid, and other malignan- Surgical resection of liver metastases still is con-
cies also are treatable. According to our inclu- sidered one of the best options for radical treat-
sion criteria, patients who are eligible for this ment of malignant tumors, but only 20% of
treatment have fewer than five lesions, a patients are candidates. Clinical conditions such
maximum diameter of 50mm, are unfit for sur- as the presence of lesions in both hepatic lobes
gical resection, do have unresectable tumors in or poor overall status of the patient excludes
both hepatic lobes, or refuse surgical resection. surgical treatment. Additional liver surgery is
Those patients who have had partial resec- associated with a mortality rate of approxi-
tion of one hepatic lobe and develop a new mately 3 to 8%.A major problem after surgical
lesion in the remaining liver also are suitable resection is tumor recurrence in up to 70% of
for this laser therapy. Requirements in cases. Only in selected cases is re-resection pos-
the latter group are complete resection of the sible, so that further surgery is untenable for
primary tumor and absence of extrahepatic most patients.
metastases. Magnetic resonance imaging-guided LITI
offers the option of multiple retreatment ses-
sions. For new hepatic lesions the same inclu-
Indications and Inclusion Criteria for sion criteria are applied as for the initial LITT.
In the literature Stangl followed up 1099
LITT of Hepatic Tumors consecutive patients with colorectal liver
• Maximum of five lesions metastases; 566 of them (51.5%) received
• Maximum diameter of 50 mm no treatment for their hepatic metastases; 340
• Patients with tumor recurrence after surgery, (31 %) underwent hepatic resection; 123
radiation, or chemotherapy (11.2 %) received regional chemotherapy; and
• New metastases after hepatic resection 70 (6.4%) received systemic chemotherapy.
• No response to chemotherapy Thirty-four patients died within 30 days, as a
• Metastases in both lobes of the liver result either of postoperative complications or
• Lesions in high-risk locations, for example, advanced disease; 48 were excluded, because
near the bile duct they developed a second primary cancer. After
• LITT as a replacement for chemotherapy if hepatic resection, 60% of these patients sur-
patient refusal vived 5 years (median survival 30 months). In
18. Percutaneous Laser Therapy 243

patients who underwent regional or systemic promlsmg for real-time monitoring of the
chemotherapy, the median survival was 12.7 progress of freezing and thawing. Hospitaliza-
and 11.1 months, respectively. In the untreated tion of about 7 to 10 days is typical with surgi-
group, 31.3% of the patients were alive after 1 cal cryotherapy.
year,7.9% after 2 years, 2.6% after 3 years, and
0.9% after 4 years. Considering these data, our Radiofrequency Ablation
results are comparable to those of surgery. The
lack of perioperative mortality and low rate Radiofrequency (RF) ablation is another
of side effects combined with an outpatient modality for thermal coagulation of tumors.
management are responsible for high patient Heating is caused by high tissue impedance
tolerance. Patients with unresectable liver between two applicator tips. Solbiati demon-
tumors need multimodality therapy for pallia- strated the feasibilty of tumor destruction with
tion, while preserving a good quality of life. conventional and cooled-tip monopolar RF
But LITI alone, or combined with systemic electrodes in patients with metastatic gastroin-
or locoregional chemotherapy, transarterial testinal carcinomas. The same inclusion criteria
chemoembolization, or surgical resection im- for LITT are used, but problems exist with
proves local tumor control and tumor-free sur- monitoring of the procedure. Interference
vival in patients with metastatic discase. between RF systems and MRI devices pre-
vents simultaneous imaging, monitoring, and
treatment.
Percutaneous Ethanol Injection
Magnetic resonance imaging-guided ethanol Focused Ultrasound
ablation is used to reduce tumor bulk and
Noninvasive therapy using focused, high-
cancer pain, but mostly in low-risk regions, and
intensity ultrasound beams was first proposed
it has been used particularly to treat HCC.
as a therapeutic modality for destruction of
Among the limitations of this agent is the lack
central nervous system tissue. Recently, a novel
of an instrument to monitor the effects in both
approach has been used for real-time monitor-
normal and pathologic tissue. Comparisons
ing of focused ultrasound using only MRI to
between interstitial laser coagulation and per-
demonstrate temperature elevation during son-
cutaneous alcohol injection to treat colorectal
ication and to delineate tissue necrosis. This
hepatic metastases have shown that there were
promising modality is under evaluation for soft
no major complications after either method,
tissue tumors in the brain and the breast. Prob-
but that pain during alcohol injection was more
lems are the rapid increase of temperature and
severe and tumor control was less. In summary,
reaching larger volumes with a small focus. So
alcohol injection is an accepted therapy method
far, focused ultrasound is not used to treat liver
for small HCC nodules, but it is not a treatment
metastases due to breathing artifacts.
option for metastases.

Transarterial Chemoembolization
Cryotherapy
Transarterial chemoembolization (TACE) is
At present, cryotherapy is carried out using defined as a local infusion of chemotherapy
laparotomy and ultrasound guidance. Single into tumor vessels to treat HCC nodules and
and multiprobe arrays are possible. Tumor neuroendocrine hepatic metastases. The goal is
tissue is frozen at temperatures approaching palliative, to reduce tumor bulk and to deceler-
-190 degrees, and then thawed. The double- ate tumor progression.
freezing technique results in reliable destruc-
tion of tumor cells. Inclusion criteria are similar
to LITT as to size and number of lesions.
Systemic and Regional Chemotherapy
Cryotherapy under MRI guidance has been All the above-mentioned techniques are
investigated in clinical studies, and MRI is restricted regionally to the liver, so the adjunc-
244 T.J. Vogi et al

tive use of systemic chemotherapy is essential enhancement along with a reliable safety
in most cases. Protocols, doses, and intervals margin.
depend on the primary tumor and data from Local tumor control rates for data from 1997
staging. to 2000 are 98.1 % after 3 months, and 97.3%
after 6 months in liver lesions smaller than 50
mm. Mean survival time for all patients is 45
Results months using the Kaplan-Meier method (95%
confidence interval: 40.9 to 49.2 months, median
In Vitro Studies 39.8 months, maximum survival 74.6 months)
In vitro studies using porcine liver demon- (Fig. 18.5).
strated a reproducible loss of signal intensity The most homogeneous group of patients are
in MRI in accordance with increasing tissue those with hepatic metastases from primary
temperature. Using an energy of 5 Wand an
application time of 12 minutes, the maximum
diameter of signal loss was 25 mm. This effect 1 ,O.--,.-------~---~------,,.---------,
was best monitored using the thermo-turbo-
FLASH sequence at TI values of 300 to 400 ms; ~ ,8 "
, . .
_ ..
this provided a nearly linear, inverse correla-
tion between signal intensity and temperature,
as well as with thermo-FLASH-2D sequences.
~ ,6 T ; .
A mean size of necrosis of 2 cm3 with an ellip-
soid morphology may be achieved by an appli-
i ,4 , r· t .
cation time of approximately 20 minutes and a o
power of 5 to 6 W using one applicator system. ,2 + ·L .

There are several possibilities to enlarge the


necrosis volume: first is the pullback technique, 0,0+--_ _~----4----....;.....--~
that allows a longitudinal enlargement of o 2 4 6 8
necrosis. Second is the multiapplicator tech- survival time [years] A
nique. The placement of two or three applica- 1,O ........-=--~-~-~-,.---------,,.-------,-~---,
tor systems results in a volume of necrosis of up
to 17 cm3 • The use of an internally cooled laser i ~
,8 _ 1 ·· ·..· _ · ·
applicator at power settings between 25 and
30 W for 10 to 20 minutes also produces signifi-
cant enlargement of the coagulative necrosis ,6 -r t j · --:- ·.._ · ·
volume.
,4 + _.. . . . . . . . . , - :- 7 ..

Patients
From 1993 to July 2002, we treated 1159
,2 7 • ·, ·+ r · ·
patients with a total of 2505 liver metastases
O,O-l--_+--_+--_+--_;...-_;...-__'--__,.--------l
from colorectal carcinoma, esophageal, gastric,
o 2 345 678
pharyngeal, testicular, and pancreatic tumors. A
total of 9470 laser applications were performed. survival time [years] B
The laser-induced necrosis was quantified by
FIGURE 18.5. (A) The mean survival time in the total
comparing the pre- and posttherapeutic unen- population of patients with liver metastases is
hanced and contrast-enhanced MRL Parame- 48.0 months (95% confidence interval: 44.4-52.8
ters such as size, morphology, and contrast months). (B) For patients with liver metastases
enhancement were compared to pretherapeutic from colorectal carcinoma the mean survival time is
MRL Successful therapy has been defined by a 41.8 months (95% confidence interval, 37.3-46.4
geometrically shaped necrotic area without months).
18. Percutaneous Laser Therapy 245

colorectal carcinoma. The mean survial rate is Fourteen patients with abdominal lymph
42.6 months (95% confidence interval: 37.7 to node tumors or recurrent primary pelvic
47.5 months, median 36.7 months). tumors were treated successfully using MRI-
Survival did not differ significantly (p > .05) guided LITI. Reduction of clinical symptoms
between men and women, or between patients was seen in 68% these patients.
with colorectal metastases and those with Magnetic resonance imaging-guided LITT
metastases from other primary tumors. allows accurate online thermometry during
these interventional procedures. Dynamic
gadolinium-enhanced MRI is suitable for
Complications early and late follow-up studies for lesions
All patients tolerated the procedure well under treated with LITT. Follow-up studies indicate
local anesthesia. The following side effects were that these laser-induced effects lead to reli-
documented by clinical examination or imaging able palliation in recurrent head and neck
studies: pleural effusion in 7.28%, subcapsular tumors.
hematoma in 2.46%, intrahepatic abscess in
0.8%, intraabdominal bleeding in 0.11 %, and
local infection at the site of the puncture in Conclusions
0.2%.
All complications except the following did Percutaneous MRI-guided interstitial laser-
not require therapy-in 0.8% of cases pleural induced thermotherapy (LITT) of malignant
effusion had to be treated by percutaneous liver tumors is a reliable treatment method to
drainage. All abscesses were treated by percu- destroy tumors palliatively, and it is also poten-
taneous drainage. Three patients died of LITT tially curative. The treatment concept should
within 4 weeks. One patient with a suspected be differentiated according to the underlying
colonic perforation had surgical repair. histology: for hepatocellular carcinoma a
local ablative procedure instead of, or in com-
bination with, local alcohol instillation or
Head and Neck and Extrahepatic transarterial chemoembolization (TACE) can
Tumors be used.
Twenty-six patients (eight women and 18 men, In the case of liver metastases, the thera-
mean age 64 years, range 57-77 years) with peutic situation must be discussed within
recurrent tumors of the head and neck were the context of the primary tumor. Today,
treated with LITT. In all these patients the using MRI-guided LITI for hepatic metastases
primary tumor was located in the head and restricted to a local area without extrahepatic
neck region. Recurrent squamous cell carci- tumor can be justified clinically. Magnetic reso-
noma occurred in the majority of patients. Two nance tomography is a valuable tool to monitor
patients with pleomorphic adenoma in the and control percutaneous LITI. Magnetic res-
parapharyngeal space were treated for tumor onance imaging also is used for follow-up. It is
recurrence after primary surgery (Figs. 18.3 and proven to be the optimal imaging examination
18.4). All patients tolerated the procedure well to detect very small tumor recurrences.
under local anesthesia. No side effects were The ultimate goal of MRI-guided LITT is
observed. 100% eradication of local tumor, as MRI online
The procedure was judged successful if thermometry allows precise guidance of the
postinterventional MRI revealed a sharply interventional procedure. Magnetic resonance
delineated necrosis that exceeded the diameter imaging provides unparalleled topographic
of the pretherapeutic tumor. Reduction of clin- accuracy due to its excellent soft tissue contrast
ical symptoms, such as pain, was observed in 11 and high spatial resolution. Therefore, early
patients. Amelioration of swallowing problems detection of local complications such as bleed-
and symptoms of nerve compression occurred ing, as well as the desired effect of coagulative
in six patients. necrosis are all possible. Dynamic contrast-
246 T.J. Vogi et al

enhanced MRI represents the optical method sis of prognostic determinants. Br I Surg 1991;
to evaluate the treated lesions, short- and long- 78:797-801.
term. Ekberg H, Tranberg KG, Andersson R, Hagerstrand
I, Ranstam I, Bengmark S. Determinants of sur-
Bibliography vival in liver resection for colorectal secondaries.
Br I Surg 1986;73:727-731.
Adson MA, Heerden van I, Adson MH, Wagner Feifel G, Schiidder G, Pistorius G. Kryochirurgie-
IS, Ilstrup DM. Resection of hepatic metastases Renaissance oder echter Fortschritt? Der Chirurg
from colorectal cancer. Arch Surg 1984;119:647- 1999;70:154-159.
651. Fortner IG, Silva IS, Golbey RG, Cox EB, Maclean
Amin Z, Bown SG, Lees WR. Local treatment of BJ. Multivariate analysis of a personal series of
colorectalliver metastases: a comparison of inter- 247 consecutive patients with liver metastases
stitial laser photocoagulation (ILP) and percuta- from colorectal cancer. Ann Surg 1984;199:306-
neous alcohol injection (PAl). Clin Radiol 1993; 315.
48(3):166-171. Fried MP, Morrison PR, Hushek SG, Kernahan GA,
Anzai Y, Desalles AA, Black KL, et a1. Interven- Iolesz FA. Dynamic T1-weighted magnetic reso-
tional MR imaging. Radiographies 1993;13(4): nance imaging of interstitial laser photocoagula-
897-904. tion in the liver: observations on in vivo
Bozzetti F, Doci R, Bignami P, Morabito A, Gennari temperature sensitivity. Lasers Surg Med 1996;
L. Patterns of failure following surgical resection 18(4):41G-419.
of colorectal cancer liver metastases. Ann Surg Gayowski TI, Iwatsuki S, Madariaga IR. Experience
1987;205(3):264-269. in hepatic resection for metastatic colorectal
Brewer WH, Austin RS, Capps GW, Neifeld IP Intra- cancer: analysis of clinical and pathological risk
operative monitoring and postoperative imaging factors. Surgery 1994;116:703-710.
of hepatic cryosurgery. Semin Surg OncoI1998;14: Gewiese B, Beuthan I, Fobbe F, et a1. Magnetic res-
129-155. onance imaging-controlled laser-induced intersti-
Butler I, Attiyeh FF, Daly 1M. Hepatic resection for tial thermotherapy. Invest Radiol 1994;29(3):345-
metastases of the colon and rectum. Surg Gynecol 351.
Obstet 1986;162(2):109-113. Goldberg SN, Gazelle GS, Solbiati L, et a1. Ablation
Cady B, Stone MD, McDermott WV, et a1. Technical of liver tumors using percutaneous RF therapy.
and biological factors in disease-free survival after AIR 1998;170:1023-1029.
hepatic resection for colorectal cancer metastases. Griffith KD, Sugarbaker PH, Chang AE. Repeat
Arch Surg 1992;127:561-569. hepatic resections for colorectal metastases.
Charnley RM, Doran I, Morris DL. Cryotherapy Surgery 1990;107:101-104.
for liver metastases: a new approach (comment Harned RK, Chezmar IL, Nelson RC. Recurrent
in Br I Surg 1990;77:354). Br I Surg 1989;76: tumor after resection of hepatic metastases from
1040. colorectal carcinoma: location and time of dis-
Cline HE, Hynynen K, Hardy CI, Watkins RD, covery as determined by CT. AIR 1994;163(1):93-
Schenck IF, Iolesz FA. MR temperature mapping 97.
of focused ultrasound surgery. Magn Reson Med Herfarth C, Heuschen UA, Lamade W, Lehnert T,
1994;31(6):628-636. Otto G. Rezidiv-Resektionen an der Leber bei
Cline HE, Hynynen K, Watkins RD, et a1. Focused primaren und sekundaren Lebermalignomen.
US system for MR imaging-guided tumor abla- Chirurg 1995;66:949-958.
tion. Radiology 1995;194(3):731-737. Hill CR. Optimum acoustic frequency for focused
Cline HE, Schenck JF, Hynynen K, Watkins RD, ultrasound surgery. Ultrasound Med BioI 1994;
Souza SP, Iolesz FA. MR-guided focused ultra- 20(3):271-277.
sound surgery. I Comput Assist Tomogr 1992; Hohenberger P, Schlag P, Schwarz V, Herfarth C.
16(6):956-965. Leberresektion bei Patienten mit Metastasen
Cline HE, Schenck IF, Watkins RD, Hynynen K, colorektaler Carcinome. Ergebnisse und prognos-
Iolesz FA. Magnetic resonance-guided thermal tische Faktoren. Chirurg 1988;59:41G-417.
surgery. Magn Reson Med 1993;30(1):98-106. Holm A, Bradley E, Aldrete IS. Hepatic resection of
Doci R, Gennari L, Bignami P, Montalto F, Bozzetti metastases from colorectal carcinoma. Morbidity,
F. One hundred patients with hepatic metastases mortality and pattern of recurrence. Ann Surg
from colorectal cancer treated by resection: analy- 1989;209(4):428-434.
18. Percutaneous Laser Therapy 247

Hughes KS, Simon R, Songhorabodi S, et al. Resec- sectable hepatic malignancies. Am Surg 1997;63:
tion of the liver for colorectal carcinoma metas- 923-926.
tases: a multi-institutional study of indications for Lorentzen T. A cooled needle electrode for radio-
resections. Surgery 1988;103(3):278-288. frequency tissue ablation: thermodynamic aspects
Hughes KS, Simon R, Songhorabodi S, et al. Resec- of improved performance compared with conven-
tion of the liver for colorectal carcinoma metas- tional needle design. Acad Radiol 1996;3(7):556-
tases: a multi-institutional study of pattern of 563.
recurrence. Surgery 1986;100(2):278-284. Lorenz M, Rossion I. Adjuvante und palliative
Iwatsuki S, Shaw BW, Starzl TE. Experience with 150 regionale Therapie von Lebermetastasen kolorek-
liver resections. Ann Surg 1983;197(3):247-253. taler Tumoren. Dtsch Med Wochenschr 1995;120:
Jolesz FA, Bleier AR, Jakab P, Ruenzel PW, Huttl K, 690-697.
Jako GJ. MR imaging of laser-tissue interactions. Lorenz M, Staib-Sebler E, Rossion I, Koch B, Gog C,
Radiology 1988;168(1):249-253. Encke A. Ergebnisse der Resektion und adjuvan-
Jolesz FA, Blumenfeld SM. Interventional use of ten Therapie von Lebermetastasen kolorektaler
magnetic resonance imaging. Magn Reson Q 1994; Primartumoren--eine Literaturtibersicht. Zen-
10(2):85-96. tralbl Chir 1995;120:769-779.
Kahn T, Bettag M, Ulrich F, et al. MRI-guided laser- Lufkin RB, Robinson JD, Castro DJ, et al. Interven-
induced interstitial thermotherapy of cerebral tional magnetic resonance imaging in the head
neoplasms. J Comput Assist Tomogr 1994;18(4): and neck. Top Magn Reson Imaging 1990;2(4):
519-532. 76-80.
Lee FT, Mahavi DM, Chosy SG, et al. Hepatic Martin M, Tarara D, Wu YM, et al. Intrahepatic
cryosurgery with intraoperative US guidance. arterial chemoembolization for hepatocellular
Radiology 1997;202:624-632. carcinoma and metastatic neuroendocrine tumors
Lee TH, Anzai Y, Lufkin RB. Magnetic resonance in the era of liver transplantation. Am Surg 1996;
imaging-guided needle biopsy of head and neck 62:724-732.
lesions. West J Med 1993;159(1):69. Masters A. Cryotherapy for liver metastases [letter;
Lencioni R, Goletti 0, Armillotta N, et al. Radio- comment]. Br J Surg 1990;77(3):354.
frequency thermal ablation of liver metastases Masters A, Steger AC, Lees WR, Walmsley KM,
with a cooled-tip electrode needle: results of a Bown Sa. Interstitial laser hyperthermia: a new
pilot clinical trial. Eur RadioI1998;8:1205-1211. approach for treating liver metastases. Br J Cancer
Lewin JS, Connell CF, Duerk JL, et al. Interactive 1992;66(3):518-522.
MRI-guided radiofrequency interstitial thermal Matsumoto R, Oshio K, Jolesz FA. Monitoring of
ablation of abdominal tumors: clinical trial for laser and freezing-induced ablation in the liver
evaluation of safety and feasibility. J Magn Reson with Tl-weighted MR imaging. J Magn Reson
Imaging 1998;8:40-47. Imaging 1992;2(5):555-562.
Livraghi T, Giorgio A, Marin G, et al. Hepatocellular McDannold N, Hynynen K, Wolf D, Wolf G, Jolesz F.
carcinoma and cirrhosis in 746 patients: long-term MRI evaluation of thermal ablation of tumors
results of percutaneous ethanol injection. Radiol- with focused ultrasound. J Magn Reson Imaging
ogy 1995;197(1):101-108. 1998;8:91-100.
Livraghi T, Goldberg N, Lazzaroni S, Meloni B, Nordlinger B, Guiguet M, Vaillant JC, et al. Surgical
Solbiati L, Gazelle GS. Small hepatocellular resection of colorectal carcinoma metastases to
carcinoma: treatment with radiofrequency abla- the liver. A prognostic scoring system to improve
tion versus ethanol injection. Radiology 1999;210: case selection, based on 1568 patients. Assoc Franc
655-661. Chirurg Cancer 1996;77(7):1254-1262.
Livraghi T, Goldberg S, Monti F, et al. Saline- Nordlinger B, Vaillant J-c. In Hepatobiliary Cancer,
enhanced radiofrequency tissue ablation in the Sugarbaker, P, editor. Repeat Resections for
treatment of liver metastases. Radiology 1997; Recurrent Colorectal Liver Metastases. Boston:
202:205-210. Kluwer Academic, 1994:57-61.
Livraghi T, Lazzaroni S, Meloni F, Torzilli G, Vettori Ohlsson B, Stenraum U, Tranberg KG. Resection
C. Intralesional ethanol in the treatment of unre- of colorectalliver metastases: 25-year experience.
sectable liver cancer. World J Surg 1995;19(6): World J Surg 1998;22:268-276.
801-806. Onik GM, Atkinson D, Zemel R, Weaver ML.
Lopez RL, Pan SH, Lois JF, et al. Transarterial Cryosurgery of liver cancer. Semin Surg Oncol
chemoembolization is a safe treatment for unre- 1993;9:309-317.
248 T.J. Vogi et al

Ravikumar TS, Kane R, Cady B, Jenkins R, Clouse colorectalliver metastases. Lancet 1994;343(8910):
M, Steele G. A 5-year study of cryosurgery in the 1405-1410.
treatment of liver tumors. Arch Surg 1991;126: Steele G Jr. Cryoablation in hepatic surgery. Semin
1520-1524. Liver Dis 1994;14(2):120-125.
Ringe B, Bechstein WO, Raab R, Meyer H-J, Steele G, Osteen RT, Wilson RE, et al. Patterns of
Pichlmayr R. Leberresektion bei 175 Patienten failure after surgical cure of large liver tumors. Am
mit colorektalen Metastasen. Chirurg 1990;61: J Surg 1984;147:554-559.
272-279. Sugihara K, Hojo K, Moriya Y, Yamasaki S, Kosuge
Roberts HRS, Paley M, Sams VR, et al. Magnetic T, Takayama T. Pattern of recurrence after hepatic
resonance imaging control of laser destruction resection for colorectal metastases. Br J Surg
of hepatic metastases: correlation with post- 1993;80(8):1032-1035.
operative helical CT. Min Invas Ther Allied van Hillegersberg R. Fundamentals of laser surgery.
Technol 1997;6:53-64. Eur J Surg 1997;163:3-12.
Rossi S, Di Stasi M, Buscarini E, et al. Percutaneous Viadana E, Bross IDJ, Pickren JW. The metastatic
RF interstitial thermal ablation in the treatment of spread of cancers of the digestive system in man.
hepatic cancer. AJR 1996;167:759-768. Oncology 1978;35:114-126.
Sanz-Altamira PM, Spence LD, Hubermann LS, Vogi TJ. Eichler K, Straub R, et al. Laser-induced
et al. Selective chemoembolization in the manage- thermotherapy of malignant liver tumors: general
ment of hepatic metastases in refractory colorec- principles. equipment, procedure, side-effects,
tal carcinoma: a phase II trial. Dis Colon Rectum complications, and results. Eur J Ultrasound
1997;40:770-775. 2001;13(2):117-127.
Sarantou T, Bilchik A, Ramming KP. Complications Vogi TJ, Mack M, Straub R, Engelmann K, Eichler
of hepatic cryosurgery. Semin Surg OncoI1998;14: K, Zangos S. Interventional MRI: interstitial
156-162. therapy. Eur RadioI1999;9:1479-1487.
Scheele J, Altendorf-Hofmann A, Stangl R, Schmidt Vogi TJ, Mack MG, Straub R, Roggan A, Felix R.
K. Surgical resection of colorectalliver metastases: Magnetic resonance imaging-guided abdominal
gold standard for solitary and completely interventional radiology: laser-induced ther-
resectable lesions. Swiss Surg Suppl 1996;4:4-17. motherapy of liver metastases. Endoscopy 1997;
Scheele J, Stangl R, Altendorf Hofmann A, Gall FP. 29:577-583.
Indicators of prognosis after hepatic resection for Vogi TJ, Mack M, Straub R, Roggan A, Felix R. Per-
colorectal secondaries. Surgery 1991;110(1):13-29. cutaneous MRI-guided laser-induced thermother-
Schlag P, Hohenberger P. Herfarth C. Resection of apy for hepatic metastases for colorectal cancer.
liver metastases in colorectal cancer~ompetitive Lancet 1997;350:29.
analysis of treatment results in synchronous versus Vogi TJ, Mack MG, Straub R, et al. MR-guided laser-
metachronous metastases. Eur J Surg Onco11990; induced thermotherapy of malignant liver lesions:
16:360-365. technique and results. Onkologie 1998;21:412-
Schwarzmaier HJ. Kahn T. Magnetic resonance 419.
imaging of microwave induced tissue heating. Vogi TJ, Muller PK, Hammerstingl R, et al. Malig-
Magn Reson Med 1995;33(5):729-731. nant liver tumors treated with MR imaging-guided
Shiina S. Tagawa K. Unama T, et al. Percutaneous laser-induced thermotherapy: technique and
ethanol injection therapy of hepatocellular carci- prospective results. Radiology 1995;196(1):257-
noma: analysis of 77 patients. AJR 1990;155: 265.
1221-1226. Vykhodtseva NI, Hynynen K, Damianou C. Pulse
Sironi S, De Cobelli F, Livraghi T, et al. Small hepa- duration and peak intensity during focused ultra-
tocellular carcinoma treated with percutaneous sound surgery: theroretical and experimental
ethanol injection: unenhanced and gadolinium- effects in rabbit brain in vivo. Ultrasound Med
enhanced MR imaging follow-up. Radiology 1994; Bioi 1994;20(9):987-1000.
192(2):407-412. Weaver ML,Ashton JG,Zemel R. Treatment of colo-
Solbiati L, Goldberg SN, Ierace T, et al. Hepatic rectal liver metastases by cryotherapy. Semin Surg
metastases: percutaneous radio-frequency abla- OncoI1998;14:163-170.
tion with cooled-tip electrodes. Radiology 1997; Weiss L, Grundmann E, Torhorst J, et al. Haematoge-
205(2):367-373. nous metastatic patterns in colonic carcinoma: an
Stangl R, Altendorf Hofmann A, Charnley RM, analysis of 1541 necropsies. J Pathol 1986;150(3):
Scheele 1. Factors influencing the natural history of 195-203.
18. Percutaneous Laser Therapy 249

Wingo PA, Tong T, Bolden S. Cancer statistics, 1995. Zhou X-D, Tang Z-Y, Yu Y-Q, et al. The role of
CA Cancer J Clin 1995;45(1):8-30. cryosurgery in the treatment of hepatic cancer: a
Yeh KA, Fortunato L, Hoffmann Jp, Eisenberg BL. report of 113 cases. J Cancer Res Clin Onco11993;
Cryosurgical ablation of hepatic metastases 120:100-102.
from colorectal carcinomas. Am Surg 1997;63(1):
63-67.
19
Cryoablation: History, Mechanism of
Action, and Guidance Modalities
Sharon M. Weber and Fred T. Lee, Jr.

History of Cryoablation widespread, and are currently still used for a


variety of skin conditions. During this time, the
Cold temperatures have been used to decrease clinical sequelae of freezing skin were well
inflammation and relieve pain since the time described in many medical journals. The clini-
of the ancient Egyptians. James Arnott (1797- cal manifestations included bulla formation,
1883), an English physician, is credited with followed by superficial crusting, then eschar
being the first to use cold to destroy tissue, formation, and finally healing to form a super-
using a combination of ice and salt to produce ficial scar.
tissue necrosis (1). Because this cryogen needed In the 1950s, Allington became one of the
to be applied topically to tumors, he was able first to recognize the importance of liquid nitro-
to treat only tumors of the cervix and breast; he gen for surgical applications. He described a
reported decreases in tumor size and palliation technique of dipping cotton swabs into liquid
of pain in patients treated by cryoablation (1). nitrogen followed by direct application to
The utility of liquid air and carbon dioxide benign skin lesions. In the late 1960s, the clini-
(C0 2) as cryogens was popularized in the late cal indications for low-temperature therapy
1890s. The use of these substances was based on expanded to treat other organ sites including
a principle that has since been used for air con- oral cancers, ophthalmologic, and gynecologic
ditioning and refrigeration-atmospheric gases conditions. Also in the 1960s, the invention of
warm when compressed and cool during expan- other devices, including a liquid nitrogen spray
sion. The first clinical application of liquid air in a hand-held form, helped popularize the use
was reported by White, who used it to treat of cryoablation for skin lesions. These devices
various skin conditions. He also described its improved the depth of penetration of the cryo-
potential curative action for carcinoma. Liquid lesion, and thus improved the efficacy for
CO 2 was first used in the early 1900s by a treatment of many skin lesions (2).
number of investigators, including Pusey, In 1963 Cooper (3), a New York neurosur-
Cranston-Low, Whitehouse, and Hall-Edwards. geon, reported on a liquid nitrogen probe
These investigators described liquid CO 2 to capable of achieving a temperature of -196°C.
treat skin conditions with minimal resultant The Cooper device was an automated system
scarring. Part of the appeal of liquid CO 2 was that utilized the continuous flow of liquid nitro-
that it was both easier to obtain and simpler to gen through a closed, self-contained cryoprobe.
maintain in liquid form than liquid air. Both This unit was one of the first to use pressurized
Pusey and Whitehouse made initial observa- liquid nitrogen pumped through a cryoprobe,
tions regarding the relationship between pres- and was capable of temperature-controlled
sure on the skin and increasing depth of regulation of the freezing process. This device
penetration (1). These techniques became fairly made possible the ability to rapidly create a

250
19. Cryoablation 251

continuous large-volume freeze in deep-seated drainage due to damage to the urethra (1). The
tissue. The probes initially were used to treat complication rate for cryoablation monitored
inoperable brain tumors and Parkinson's by physical examination was high enough that
disease by focally ablating affected areas in the the technique rapidly lost favor, and was only
brain. revived in the mid-1980s.
The novel idea of using liquid nitrogen to The key development that sparked renewed
cool a probe for placement into less accessible interest in cryoablation was the fusing of
body spaces was instrumental in ushering in the cryoablation with real-time imaging guidance
modern era of cryoablation and increasing the in the form of intraoperative ultrasound (4,5).
number and variety of applications. Up to this Liver tumors were the first cancer to be
point, freezing of tissue had been limited to approached with this method. During the early
easily accessible organs and small volumes of to mid-1980s, surgeons had become increas-
tissue. Development of liquid nitrogen cryo- ingly aware of the segmental anatomy of the
probes dramatically augmented the volume of liver, and the importance of the blood supply to
tissue that could be frozen by low probe tem- individual hepatic segments (6). Knowledge of
peratures, and increased the types of tissues this anatomy allowed hepatic surgeons to resect
that could be ablated. The continued develop- liver tumors more safely and aggressively. As a
ment of systems based on this technology has result, the morbidity and mortality from hepatic
helped to broaden the clinical applications of surgery have greatly decreased during the past
cryoablation. In the 1970s and 1980s, several two decades, accompanied by a concomitant
other indications for cryoablation using liquid increase in overall and disease-free survival (7).
nitrogen systems were investigated, including During this period of increased interest in
ablation of prostate, kidney, and liver cancers. hepatic surgery, intraoperative ultrasound was
Despite the increased interest and investiga- discovered to be an excellent tool to define liver
tion into tumor ablation by low temperatures, anatomy for resection and to evaluate the liver
cryoablation of deep-seated thoracoabdominal for the presence of occult metastatic disease.
and pelvic tumors did not become a clinically Intraoperative ultrasound is now widely con-
important tool until the 1980s. The main reason sidered to be the gold standard for imaging the
for this was simple-physicians could not ade- liver (8-10).
quately control the extent of freezing and tissue It was Onik et aI's (4,5) discovery of the
death caused by powerful cryoprobes that highly echogenic nature of the iceball that
used liquid nitrogen. Several clinical series forms during hepatic cryoablation that sparked
demonstrated the ability to kill targeted tissue renewed interest in hepatic cryoablation and
effectively, but the methods for monitoring ultimately led to the development of other
the freeze were crude at best. In most cases, thermal ablation devices and techniques. Sub-
the freeze was controlled by inspection and sequent animal studies showed a close correla-
palpation of the iceball. In other words, the tion between the sonographically visible iceball
physician had to rely primarily on physical and the zone of cell death (11,12). The ability
examination to determine whether the targeted to monitor the formation of the iceball pre-
tissue had been adequately treated without cisely helped overcome the primary limitation
inadvertently freezing nontargeted tissue. The of cryoablation to that time-the uncontrolled
results of cryoablation monitored by physical and poorly monitored freezing process. With
examination were predictably mixed. Most intraoperative ultrasound monitoring, cry-
reports during this period showed some efficacy oprobes could now be placed precisely with
in terms of tumor kilL but severe complica- real-time guidance into the tumor, thus
tions also were common. Tissue vulnerable decreasing injury to hepatic vessels and bile
to cryoablation (bile ducts, urethra, bowel, ducts by the probes. The cryolesion also could
etc.) often was irreparably damaged (1). For be controlled well enough to allow for freezing
instance, in prostate cryotherapy, these compli- of the targeted tumor and a margin of normal
cations often resulted in prolonged catheter liver, while decreasing the frequency of injuries
252 S.M. Weber and F.T. Lee, Jr.

to the bile ducts and extrahepatic structures. is local tissue ischemia due to small-vessel
This process has become more precise over the thrombosis (26,27).
ensuing decades, and many reports of low com- To destroy tumor cells, it has become clear
plication rates coupled with complete tumor that temperatures must reach a certain critical
kill in a high percentage of cases are now threshold that is unique to the cell type and
common in the scientific literature (13-16). thermal environment of the targeted tissue.
While initially developed to monitor hepatic Estimates of the critical temperature for com-
cryoablation, Onik et al (4,17) and others plete tissue destruction range between -20 and
(18,19) have since used intraoperative ultra- -50°C, depending on the tissue. In addition,
sound in combination with cryoablation to treat studies on isolated cell suspensions have shown
prostate, renal, and various other benign and that multiple freeze cycles, an increased rate of
malignant conditions. freezing, and colder temperatures all contribute
The next technical breakthrough that to a decrease in viable cells after cryoablation
prompted further development of cryoablation (28-30).
techniques was the introduction of argon gas There is experimental evidence that suggests
systems based on the Joule-Thompson prin- that a single freeze-thaw cycle results in
ciple (20). These systems increased the con- complete cell death in large animal models;
venience of freezing, decreased the procedure however, these models tested cryoablation in
time, and allowed for smaller (some as small as normal tissue, not tumor (27,31). When cryoab-
17-gauge), yet highly effective cryoprobes. Most lation has been tested in small animal tumor
recently, the small probes used with argon models, it appears that multiple freeze-thaw
systems have led to the development of mini- cycles result in a greater likelihood of complete
mally invasive cryoablation techniques that can cell death. It is likely that because tumors
be performed through small incisions, laparo- are more fibrotic than normal tissue, they are
scopic ports, or percutaneously under cross- more resistant to freezing. This has been
sectional image guidance (4,18,21). shown in small animal tumor models in which
viable cells survived after a single freeze cycle
(29,30). In these subcutaneously implanted
Mechanism of Cell Destruction tumors, repetitive freeze-thaw cycles resulted
by Cold Temperature in improved rates of local tumor control (30).
Clinically, the standard treatment for patients
In the past 50 years, the biologic effects of freez- consists of two freeze-thaw cycles.
ing have been studied, primarily using liquid Different cell types are relatively more or
nitrogen-based systems. During freezing, ice less sensitive to cold than others. Melanocytes
crystals form within the intra- and extracellular are one of the most sensitive, which accounts
spaces. Intracellular ice crystal formation leads for the depigmentation of skin after freezing
to cell death by injury to either the cellular (32). On the other hand, collagen and elastin
membrane or structures within the cell, or both fibers are some of the most resilient (33). A
(22). However, intracellular ice forms only with study by Gage and colleagues (34), in which
freezing that occurs rapidly or freezing to large arteries in dogs were isolated and liquid
extremely low temperatures (22). Extracellular nitrogen was passed through them for 10
ice forms when freezing is slower, and causes minutes, first reported this observation. The
cell death by creating an osmotic gradient authors showed that after cryoablation, fol-
that results in intracellular fluid moving into lowed by normal blood flow through the arter-
the extracellular space; this results in dehydra- ies, there were no episodes of wall rupture.
tion and subsequent cell death (22-24). It also Although there were some minor histologic
has been hypothesized that extracellular ice changes in the vessel wall 2 to 4 weeks after
crystals cause deformation and rupture of the freezing, there was no change in function, either
cell membrane itself (25). Another important in the early or late postoperative period.
mechanism of cell death after cryoablation Modern studies have redemonstrated the
19. Cryoablation 253

ability of vessel walls to withstand freezing, several minutes, during which the probe needed
although tissue up to the vessel wall is ablated to be thawed, repositioned, and refrozen. In
if the temperature is cold enough (27). Com- addition, the relatively large probe sizes were
puted tomography (CT) scans following cryo- generally placed using a multistep Seldinger
ablation of liver tissue routinely demonstrate technique that could be complicated and time-
patent vessels coursing through the area of consuming. During the late 1980s and early
tissue necrosis, even in the presence of com- 1990s, liver and prostate tumors were the two
plete ablation of the tumor. Unfortunately, the main indications for cryoablation. However, the
bile ducts, urethra, bowel, and ureter are not as length and complexity of the procedures and
immune to the effects of cryoablation. Freezing the awkward, difficult-to-use equipment as well
of these structures can result in necrosis and as the requirement for high-quality intraopera-
infarction after freezing, often leading to stric- tive ultrasound discouraged widespread use.
ture, sloughing, and perforation (1,34,35). This Several studies were published in which expe-
clearly limits the use of cryoablation in certain rienced liver and prostate surgeons were
anatomic areas, unless warming of the vulner- unable to successfully perform these proce-
able structure can be accomplished. Urethral, dures without serious complications or failure
ureteral, and bile duct warming during cryoab- to ablate tumors completely (36,37).
lation all have been described as techniques to The development of argon gas-based
decrease the damage to these structures during systems based on the Joule-Thompson princi-
cryoablation (1,26). ple in the early 1990s represented a major
advance in cryoablation. Argon systems have
largely replaced liquid nitrogen units. The
Cryoablation Equipment Joule-Thompson principle states that low tem-
peratures are created by the rapid expansion of
Between the 1960s and early 1990s, liquid high pressure inert gas. The probe tips in argon
nitrogen-based systems were the only com- gas-based systems reach a temperature of
mercially available cryoablation devices in the approximately -150°C. Temperatures at the
United States. The most recent manufacturers probe tips of argon systems may appear warmer
of these devices were Cryomedical Sciences than those achieved by liquid nitrogen devices
(Accuprobe, Rockville, MD) and Candela (-196°C), but much of this temperature dis-
Corporation (Cryotech, Wayland, MA). These crepancy can be explained by the difference in
devices were based on the circulation of liquid location at which the temperatures are mea-
nitrogen through probes of varying sizes [3-8 sured. For example, in one argon-based system,
mm outer diameter (aD)]. Up to five probes temperatures are measured at the probe tip.
could be employed during a single freeze. While This is in contrast to the most popular liquid
highly effective in freezing tissue, these devices nitrogen system in which temperatures are
were quite large, and required large stainless measured as the nitrogen enters the probe and
steel dewars to store the liquid nitrogen, as well is not yet susceptible to the warming effects of
as large insulated probe cables. The dewars and tissue. When tip temperatures are measured
fill tanks were prone to leakage, and therefore and compared directly, they are similar, regard-
liquid nitrogen could not be stored for more less of the type of system (20,38). Therefore, this
than a few days at a time. Consequently, it was difference is likely not clinically significant.
difficult to perform cryoablation on short There are two manufacturers of argon-based
notice, a major disadvantage when unexpected cryoablation systems in the United States
tumors were encountered during liver surgery. (CRYOcare™, EndoCare Inc., Irvine, CA,
Other disadvantages of liquid nitrogen-based and CryoHit®TM, Seed Net™, Galil Medical,
systems included the relatively slow formation Wallingford, CT). The advantages of these
of the iceball, as well as slow switching between systems over liquid nitrogen units primarily are
the freeze and thaw modes. As a result, reposi- related to the use of high-pressure argon gas as
tioning of a frozen cryoprobe could take a cryogen. Because of the relatively low viscos-
254 S.M. Weber and ET. Lee, Jr.

ity of argon gas compared to nitrogen, it can Imaging Guidance for


be circulated very rapidly and at high pressure
through small probes (currently down to 17- Cryoablation
gauge). As a result, iceball formation com-
mences very rapidly after the initiation of the There are three main cross-sectional imaging
freezing process. Thawing can also be started techniques that are used to guide cryoablation:
and stopped rapidly which allows for much ultrasound (percutaneous or intraoperative),
more rapid repositioning of cryoprobes. Low CT (percutaneous), and MRI (percutaneous).
temperatures are reached more quickly than These will each be considered separately.
nitrogen-based systems, which is important
since in vivo studies suggest that rapid cooling
Ultrasound Guidance for
may improve tissue destruction (28). In both
Cryoablation
systems, multiple probes can be used simulta- Intraoperative ultrasound to guide cryoabla-
neously to ablate larger lesions or lesions in tion was first described by Onik in 1984 (5). This
different locations (CRYOcare™: eight probes seminal observation is largely responsible for
from 2.4-4.9mm OD, Seed Net™: up to fifteen the entire field of focal tumor ablation that
17-gauge probes, or eight 3.0mm probes). Both began with intraoperative cryoablation, and has
manufacturers make sharp-tipped cryoprobes since expanded to include many percutaneous
that can be directly placed into tumors. A mod- thermal ablation techniques. Since the original
ified Seldinger-like device (Fast-Trak™, Endo- use of ultrasound, this method has become the
Care, Irvine, CA) has also become available dominant modality to guide cryoablation of
recently that speeds probe placement com- the liver and prostate. The main advantages of
pared to first-generation Seldinger systems. The ultrasound for cryoablation are the real-time
Galil system is the only cryoablation unit that capabilities to safely guide probe placement,
has MRI compatible cryoprobes. the high spatial resolution of intraoperative
An important aspect of both cryoablation ultrasound, the highly visible nature of the ice-
systems available in the United States is the ball, and the excellent correlation between the
ability to use multiple cryoprobes and to adjust location of the iceball and the zone of cell death
the argon flow rate to "sculpt" the iceball to a (11,12).
precise shape. This is particularly important in The ability to place cryoprobes under real-
prostate cryoablation in which it is often neces- time ultrasound guidance helps to assure that
sary to create a conglomerate iceball using six the probe does not cross major vessels, bile
to eight 3.0mm cryoprobes (or up to fifteen 17- ducts, or other vulnerable structures. In addi-
gauge probes) to adequately ablate a prostate tion, the ability to correct the path of the probe
gland of asymmetric shape. Prostate cryoabla- rapidly helps minimize procedure time and the
tion often is monitored by both transrectal number of passes through the tumor. This may
biplane ultrasound and thermosensors placed in help to decrease the risk of tumor seeding. For
critical areas (Denonvillier's fascia, neurovas- liver cryoablation, many different ultrasound
cular bundles) (39). Depending on the location probes can be used to guide probe placement
of the tumor, it may be important to assure a and to monitor the iceball. The initial descrip-
hard freeze of at least -40°C in certain locations tion of intraoperative ultrasound guidance for
as determined by thermosensors. An important cryoablation used T-shaped side-fire transduc-
advantage of cryoablation over single probe ers that display the liver parenchyma from the
thermal ablation devices (specifically radiofre- liver surface (Fig. 19.1). This side-fire configu-
quency ablation) is the ability to incrementally ration minimizes interference by probe cords
adjust the amount of flow to specific probes (or during scanning of the liver. This is particularly
place an additional probe in an area that is not important because of the small space between
reaching critical temperatures). This results in a the diaphragm and liver surface into which
high degree of control over the eventual shape intraoperative probes must fit.
of the zone of necrosis that is not possible with These high-frequency (5-12MHz) transduc-
other thermal ablation techniques. ers come in linear and curvilinear probe faces,
19. Cryoablation 255

FIGURE 19.1. A small liver tumor is seen with intraoperative


ultrasound (A). After probe placement (B, arrows represent
probes), the two cryolesions growing from the two separate
probes can be seen as hyperechoic foci (C, D, E, arrows). When
the iceballs fuse, a single conglomerate iceball completely
obscures the tumor (E).
256 S.M. Weber and ET. Lee, Jr.

and remain the most widely used transducers vents the transmission of sound (Fig. 19.1). As a
for hepatic cryoablation. Other transducers result, no information can be obtained from
frequently used for liver cryoablation include structures that are engulfed by the iceball, and
an I-shaped side-fire transducer in a sagittal acoustic shadowing limits visualization poste-
configuration for longitudinal imaging, laparo- rior to the cryolesion. This can be a serious
scopic ultrasound transducers, and transvagi- problem during prostate cryotherapy when the
nallendorectal end-fire probes that can be used iceball is monitored strictly from a transrectal
for open or cryoablation performed via a approach posterior to the iceball. Once freezing
minilaparotomy. For the latter, a small incision of the posterior probes have commenced, a lack
can be made in the anterior abdominal wall of ultrasound information concerning the ante-
through which a transvaginal or endorectal rior probes results. The use of thermosensors in
transducer can be introduced, with the cryo- areas not well visualized by ultrasound has
probe directly mounted on the ultrasound become an important method to assure proper
transducer (21). Punctures of left lobe or ante- size of the zone of necrosis, particularly in
rior right lobe tumors can then be performed prostate cryoablation when it is often not pos-
precisely using an electronic puncture guide. sible to create a large ablative margin. Temper-
This approach is simpler and faster than using atures of -40°C and below at a thermosensor
a laparoscopic ultrasound transducer to guide can substantially increase confidence that tissue
laparoscopic cryoablation (Fig. 19.2). at that specific location has been ablated (28).
For prostate cryoablation, a transrectal, For liver cryoablation performed at open
biplane ultrasound transducer is mandatory. surgery, unlike prostate cryoablation, it is often
Due to the need for true orthogonal imaging, possible to monitor the iceball from more than
an end-fire transducer is not adequate. one surface. When this is not feasible because of
Under ultrasound visualization, the cryole- adhesions or other anatomic limitations, it is
sion is a highly echogenic structure that pre- important to place the cryoprobes in such a

A
FIGURE 19.2. (A) Minilaparotomy using a direct technique of conventional laparoscopic guidance
puncture technique. The cryoprobe is directly placed of cryoablation (A, right panel). For laparoscopic
on a transvaginal or transrectal ultrasound trans- ultrasound-guided cryoablation, the cryoprobe and
ducer (B). This allows a direct puncture of an ante- laparoscopic ultrasound transducer are not in a fixed
rior liver lesion using an electronic biopsy guide (A, plane, thus making it difficult to simultaneously
left panel). This is in contrast to the more difficult image the target, cryoprobe, and probe path.
19. Cryoablation 257

manner as to ensure that the deep margin is iceball (Fig. 19.3). This can lead to the spurious
adequately frozen because this margin will not assignment of echoes within the iceball.
be monitored easily from the anterior liver Mirror image artifacts also can be encoun-
surface. In our practice, because of this problem, tered when imaging the iceball, similar to the
we place more probes closer to the deep margin artifact encountered when imaging the dia-
to ensure an adequate enlation in this area. phragm or other highly echogenic structures.
Several troublesome ultrasound artifacts can When the freezing process is complete and
be encountered during ultrasound-monitored the cryolesion has been thawed, it is often pos-
cryoablation. Depending on the type of ultra- sible to visualize the extent of the zone of
sound transducer used, a "critical angle effect" necrosis. The area where the iceball was located
can be present at the edge of the iceball that becomes less echogenic than the surrounding
can increase the apparent size of the iceball tissue, and thus ablative margins can be deter-
(35). This is due to the speed of sound differ- mined (Fig. 19.4). This effect is particularly pro-
ences between soft tissue and ice that causes nounced in the liver where adjacent unablated
refraction of the sound beam. This artifact is tissue allows for comparison.
exacerbated when using vector transducers, and
is lessened with linear transducers that mini-
mize the amount of sound that strikes the Computed Tomography
iceball at oblique angles. It is important for Guidance of Cryoablation
the physician to understand this artifact and
adjust when necessary. An additional artifact The development of cryoprobes suitable for
that often is encountered during ultrasound- percutaneous use has spurred the development
monitored cryoablation is a reverberation arti- of CT-guided cryoablation. To date, cr has
fact from the highly echogenic surface of the been used to guide percutaneous cryoablation
of liver, kidney, lung, and body wall masses
(18,19,39-42). Prostate cryoablation has not yet
been performed with cr guidance due to the
requirement for the patient to be in the lithot-
omy position for the transperineal placement of
cryoprobes.
Computed tomography has several features
that make it a feasible, and in some ways a
nearly ideal, method to guide and monitor per-
cutaneous cryoablation of accessible tumors.
The first important feature of CT guidance for
cryoablation is that it is often the imaging
modality that was used initially to detect the
tumor. Thus, the physician performing the pro-
cedure can have confidence that the targeted
area is indeed what was seen on the initial CT
scan. In many cases, CT is used to follow the
patient postablation (43), and correlation of the
intraprocedural images with follow-up scans
can be extremely helpfuL Computed tomogra-
FIGURE 19.3. Reverberation artifact during liver phy also can image most structures (or the
cryoablation. Note the spurious assignment of anatomic region where the structure is located)
echoes within the iceball caused by reverberation that might be injured by the freezing process,
between the transducer face and the highly reflective namely, bowel, central bile ducts, and nerves.
iceball (arrow). (Case courtesy of Michael Ledwidge, Computed tomography scanners are also avail-
BS, ROMS, RVT, Madison, WI.) able in virtually every radiology practice in the
258 S.M. Weber and ET. Lee, Jr.

FIGURE 19.4. Metastatic neuroendocrine metastases in segments


1,5, and 8 (A). The tumor in segment 1 (calipers) is visualized
anterior to the inferior vena cava (B), and a cryoprobe (arrows)
has been placed in it using intraoperative ultrasound guidance
(C). After freezing, a hypoechoic zone is seen (D, arrows) sur-
rounding the tumor. This zone corresponds to the region of tissue
death.

United States, and radiologists are comfortable probe sizes associated with cryoablation, either
with CT-guided punctures for biopsy or fluid a large number of intermediate monitoring
drainage. Thus, the expertise needed to perform scans were needed, or the procedure needed to
CT-guided cryoablation is widespread. For be done in several steps using the Seldinger
these reasons, combined with a growing under- technique. Both of these techniques could
standing of the nearly painless nature of the potentially increase the accuracy of probe
procedure (44), the number of cryoablation placement, but came at the cost of lengthening
cases performed under CT guidance is expected the procedure time and increasing radiation
to grow dramatically in the coming years. exposure. Some CT-guided procedures could
Until recently, CT-guided procedures were take several hours with this method.
handicapped by the lack of true real-time guid- The development of CT fluoroscopy has
ance capability. Probes or needles were placed, helped increase the acceptance of CT-guided
and intermittent scans helped determine the cryoablation (45). Intermittent scanning can
post facto location of the device. This could substantially decrease the time needed for
result in the unfortunate placement of needles checking probe position. In addition, it is even
in undesirable locations, potentially increasing possible to perform nearly real-time imaging of
the complication rate. Because of the large probe placement. However, this requires con-
19. Cryoablation 259

tinuous fluoroscopy, and the radiation dose to noncontrast CT, so an exact ablative margin can
the patient and operator can be quite high. With be determined. If necessary, the patient can
CT fluoroscopy, it is possible to directly and receive a small bolus of contrast material to
rapidly place sharp cryoprobes by the trocar help visualize tumors seen only by contrast-
rather than Seldinger technique. The operator enhanced scans. When cryoprobes of 3.0mm
simply makes several quick checks to confirm OD or larger are used, streak artifact can
proper probe trajectory during placement. The degrade image quality. However, in most cases
use of CT fluoroscopy, therefore, can substan- this artifact does not substantially interfere
tially decrease procedure time. with the monitoring of freezing because of the
Perhaps the most important reason that CT highly visible iceball. This streak artifact is
is becoming an increasingly important modality much less severe when using 2.0-mm probes.
to guide and monitor cryoablation is that the Artifact can also be minimized by pulling back
iceball is seen very well when compared to soft the cryoprobes just prior to CT scanning.
tissue, and the visible iceball has a good corre- After the freezing process has finished and
lation in relation to the eventual zone of cell the iceball has melted, it is still possible to
death (43,46,47). In most cases, the iceball in see the frozen zone due to its decreased atten-
liver, kidney, or muscle measures approxi- uation compared to unfrozen tissue. This is
mately a Hounsfield units (HU). As a result, an analogous to the zone of decreased echogenic-
approximately 50 to 70 HU difference between ity of thawed tissue seen by ultrasound. After
the iceball and soft tissue makes it simple to the administration of contrast material, this
determine the extent of the freezing process zone does not enhance, and therefore post-
using CT. In many cases, the tumor and iceball contrast scans can be an excellent method to
margin both are well seen (Fig. 19.5), even with determine the exact area of tissue destruction
postcryoablation.

Magnetic Resonance Imaging


(MRI) Guidance of
Cryoablation
Like CT-guided cryoablation, the development
of MRI-guided cryoablation was spurred by the
development of small-diameter cryoprobes
suitable for percutaneous use. To date, only one
manufacturer of cryoablation equipment (Galil
Medical, Wallingford, CT) produces MRI-
compatible cryoprobes.
Percutaneous MRI-guided cryoablation
cannot be performed routinely with many
clinical MRI scanners (Fig. 19.6). Dedicated
midfield interventional MRI systems with a
FIGURE 19.5. Computed tomography (CT)-guided vertical magnet orientation are the best systems
cryoablation of the liver: Three cryoprobes have
for percutaneous cryoablation (48,49). These
been percutaneously placed in a peripheral liver
lesion under CT fluoroscopic guidance. Note the have the advantage of letting the operator
highly visible nature of the iceball compared to back- stand immediately adjacent to the patient dur-
ground liver. The arrow denotes a balloon inflated ing the procedure. However, cryoablation
peripheral to the iceball to protect the chest wall also can be performed with somewhat more
from freezing damage. (Case courtesy of Peter difficulty in magnets with "open" horizontal
Littrup, MD, Detroit. MI.) architecture.
260 S.M. Weber and ET. Lee, Jr.

A c

B o
FIGURE 19.6. Percutaneous magnetic resonance into the tumor (arrow). (C) During the freezing
imaging (MRI)-guided ablation of a renal cell carci- process, the iceball (curved arrows) is low signal
noma. (A) An oblique axial projection demonstrates intensity, and is seen to engulf the tumor in both the
a renal mass originating in the upper pole of the right axial and sagittal (D) projections. (Case courtesy of
kidney (arrow). (B) Sagittal image. Under real-time Stuart Silverman, MD, Boston, MA.)
MRI guidance, a cryoprobe (arrowheads) is placed

The rationale to use MRI for guidance and only with intravenous contrast administration.
monitoring of cryoablation stems from the fact Because of the rapid circulation time of iodi-
that MRI has a very high sensitivity for the nated contrast materials for CT, the tumor may
detection of most tumors amenable to cryoab- be detectable only transiently, limiting the
lation. Compared to CT scanning without intra- ability to guide a probe into a liver tumor under
venous contrast, it is likely that most tumors real-time guidance.
will be more visible on noncontrast MRI due to In addition to tumor visualization, ice is
the high inherent contrast resolution of MRI highly visible by MRI. In general, ice is of low
(50). This is a substantial advantage compared signal intensity on Tl-weighted images, with a
to CT, in which many tumors are detectable temperature dep~ndence to the signal intensity
19. Cryoablation 261

(51).As a result, MRI can be used to determine temic response ("cryoshock") than is present
temperature in tissue during the freezing with tissue destruction by heat. The hypothesis
process (52). is that intact cellular elements are more readily
Several other advantages of MRI guidance delivered into the bloodstream by freezing
for cryoablation have become evident with than with heat ablation, and this can result
increasing clinical experience. The nearly real- in thrombocytopenia and hepatic and renal
time ability to guide cryoprobes into virtually failure in severe cases (42). The actual incidence
any space of the abdomen is similar to that of
CT fluoroscopy (48). However, the multiplanar
capability of MRI represents a substantial
advantage over CT for both placing probes and
monitoring the freezing process.

Comparison with Other Focal


Ablative Techniques
Thermal ablation techniques cause tissue
destruction by creating ionic agitation [in the
case of radiofrequency (RF) and microwave
ablation] and heat, which results in tissue
boiling and the creation of water vapor. If lethal
temperatures are reached, protein denaturation
and vascular thrombosis result. A zone of
partial tissue destruction up to 8 mm in diame-
ter is seen surrounding the thermal lesion. The
mechanism of tissue destruction by heat is dif-
ferent from that created by cryoablation. In
cryoablation, the freezing and thawing process
destroys cell membranes and organelles due to
the mechanical stresses associated with the
phase change from ice formation. At gross
pathology, this results in a well-defined zone of
tissue destruction (Fig. 19.7).
Secondary vascular thrombosis can result,
adding to the eventual tissue death, but it is not
as prominent a feature as with the heat-based
ablation modalities. Because the cell death due
to cryoablation is primarily mechanical, cell
destruction can be readily appreciated immedi-
ately postfreeze at the light microscopic level.
Cell membranes are destroyed and cells are
clearly seen as nonviable. This differs from the FIGURE 19.7. Pathology of radiofrequency (RF) vs.
immediate postablation appearance of the cryoablation. (A) Radiofrequency ablation lesion in
normal pig liver demonstrates a central pale zone
heat-based modalities in which the cells appear
corresponding to the area of coagulation necrosis.
nearly normal, with some subtle changes in The surrounding red zone (arrows) represents
the nucleus and organelles (Fig. 19.8). Some an area of liver injury with some remaining
authors feel that the rapid destruction of viable tissue. (B) Cryoablation lesion in normal pig
cell membranes and the relative lack of liver. Note the abrupt transition between normal
protein denaturation associated with freezing and necrotic liver, and the lack of a zone of partial
is responsible for a more severe form of sys- necrosis.
262 S.M. Weber and ET. Lee, Jr.

have the iceball-air interface, is not performed


in a low-pressure environment, and has the
benefit of surrounding tissues for tamponade
(37,47,49).
The phase change seen with cryoablation
proceeds along a smoother and more pre-
dictable front than the tissue heating associated
with RF and microwave ablation. This leads to
smooth, regular-appearing, reproducible zones
of freezing during cryoablation (Fig. 19.7).
Cryoablation lesions are almost always round
FIGURE 19.8. Cryoablation, histology. Cryolesion 24
or oval in appearance. There is little deviation
hours after ablation reveals marked disruption of of the iceball by various tissue types, with the
cellular integrity with absence of cell membranes. No exception of vascular structures. Similar to
viable cells are present within the area of the freeze. heat-based ablation, patent blood vessels limit
An abrupt transition between nonviable (left, star) the ability to cause tissue necrosis because of
and viable tissue (right) is present, bordered by a thin the constant inflow of relatively warm blood.
zone of inflammation (arrow). Thus viable tumor cells may be preserved next
to blood vessels (Fig. 19.9), unless additional
attempts to achieve complete freezing are uti-
of cryoshock has probably been overestimated lized, such as using multiple probes. However,
in many reports, as a review of the world liter- because of the ability to freeze several probes
ature of cryoablation found a 3% incidence of simultaneously, vascular warming often can be
major complications (53). overcome, and tissue can be ablated up to large
Over time, postcryoablation tissue breaks blood vessels (portal vein, inferior vena cava) if
down more rapidly than tissue ablated by heat the probes are clustered and placed close
due to the existence of patent blood vessels in enough to the heat source (Fig. 19.10).
the cryoablation site. This exposes the necrotic Comparing the relative effectiveness of
tissue to inflammatory mediators and scavenger tissue destruction between RF and cryoabla-
cells of the reticuloendothelial cell system (42).
Heat-based modalities cause vascular throm-
bosis, and thus the zone of necrosis is more
avascular than with cryoablation. Tissue sub-
jected to heat ablation therefore takes a longer
time to undergo resorption and fibrosis than
cryoablated tissue.
Heat-based ablation modalities cause pro-
found vascular thrombosis. In fact, the first RF
ablation device was a modification of the Bovie
electrocautery unit (54). As a result, bleeding is
an unusual complication of RF ablation. In con-
trast, cryoablation has minimal intrinsic hemo-
static properties, and has been associated with
substantial hemorrhage during large-volume
FIGURE 19.9. Postcryoablation viable tumor (arrows)
freezes performed at open laparotomy (53). surrounding a large patent blood vessel (star) in this
In general, this is a result of cracking of the rabbit VX2 animal model. Thmor in proximity to
iceball during thawing. However, percutaneous patent blood vessels often is preserved due to the
cryoablation does not appear to have a high inflow of warm blood. Ablating tumor near large
bleeding rate, perhaps because in contrast to blood vessels is a problem for all thermal-based abla-
laparotomy, percutaneous ablation does not tion modalities.
19. Cryoablation 263

Summary
Modern cryoablation techniques have been
widely used for the focal destruction of various
different types of tissue for more than 20 years.
Cryoablation was the modality that ushered in
the modern era of focal ablation of malignant
tumors under real-time cross-sectional imaging
guidance. The advantages of cryoablation over
other thermal ablation techniques include the
ability to use multiple cryoprobes simultane-
ously; the ease of monitoring the formation of
FIGURE 19.10. Cryoablation up to a vessel wall. ice with ultrasound, CT, and MRI; the repro-
Cryoablation in normal porcine liver demonstrates ducible and predictable pattern of tissue
the ability to ablate tissue immediately adjacent to a destruction; and the relatively low complication
vessel wall when the probes are placed in proximity.
rates. Disadvantages compared with heat-based
In this case, there is an abrupt transition (arrows)
between normal liver (stars) and completely necrotic modalities include the greater systemic effects
liver. Note the complete ablation of tissue up to the seen acutely due to the lysis of cell membranes,
vessel wall (curved arrow) with preservation of the and the heretofore reluctance to use cryoabla-
patent portal vein. tion in a percutaneous fashion due to the rela-
tively large probe sizes and theoretical risk of
uncontrollable bleeding. The risk of bleeding
during percutaneous use appears to have been
overestimated, and emerging percutaneous
tion is problematic, due to the lack of well- applications are being actively investigated.
controlled studies. Most authors conclude that
cryoablation has a slight advantage in the
ability to cause cell death when tissue has been
appropriately targeted in the laboratory (55), References
but appropriately controlled clinical studies
have not been performed. In clinical trials of 1. Gage AA. History of cryosurgery. Semin Surg
hepatic tumor ablation, local recurrence rates OncoI1998;14:99-109.
generally are lower with cryoablation; these 2. Cooper SM, Dawber RP. The history of
cryosurgery. J R Soc Med 2001;94:196-201.
studies typically were performed during open 3. Cooper IS. A new method of destruction or extir-
laparotomy, and most RF ablation studies were pation of benign or malignant tissues. N Engl J
performed percutaneously, or a mix of intraop- Med 1963;263:741-749.
erative and percutaneous cases. The RF trial 4. Onik G, Gilbert J, Hoddick W, et al. Sonographic
with the lowest local recurrence rates included monitoring of hepatic cryosurgery in an experi-
a large proportion of patients with lesions that mental animal model. AIR 1985;144:1043-1047.
were ablated intraoperatively, and many with a 5. Onik G, Cooper C, Goldberg HI, Moss AA,
concurrent Pringle maneuver (56). Rubinsky B, Christianson M. Ultrasonic char-
Monitoring of tissue undergoing freezing acteristics of frozen liver. Cryobiology 1984;21:
currently is more reliable than monitoring of 321-328.
tissue undergoing heating, regardless of the 6. Fortner JG, Blumgart LH. A historic perspective
of liver surgery for tumors at the end of the mil-
imaging modality (CT, ultrasound, MRI), as dis- lennium. J Am CoIl Surg 2001;193:210-222.
cussed above. This is an important advantage of 7. Fong Y, Fortner J, Sun RL, Brennan MF, Blum-
cryoablation, as the success of any ablation gart LH. Clinical score for predicting recurrence
technique is dependent on the likelihood to after hepatic resection for metastatic colorectal
visualize the complete destruction of the tar- cancer: analysis of 1001 consecutive cases. Ann
geted tumor. Surg 1999;230:309-318.
264 S.M. Weber and ET. Lee, Jr.

8. Takigawa Y, Sugawara Y, Yamamoto J, et al. New arotomy in a porcine model-an alternative to


lesions detected by intraoperative ultrasound open cryosurgery. Surg Endosc Ultrasound
during liver resection for hepatocellular carci- Intervent Tech 1999;13:253-259.
noma. Ultrasound Med Bioi 2001;27:151-156. 22. Mazur P. The role of intracellular freezing in the
9. Bloed W, van Leeuwen MS, Borel RI. Role of death of cells cooled at supraoptimal rates. Cryo-
intraoperative ultrasound of the liver with im- biology 1977;14:251-272.
proved preoperative hepatic imaging. Eur J Surg 23. Gill W, Fraser 1. A look at cryosurgery. Scot Med
2000;166:691-(j95. J 1968;13:268-273.
10. Cervone A, Sardi A, Conaway GL. Intra- 24. Whittaker DK. Mechanisms of tissue destruction
operative ultrasound (IOUS) is essential in the following cryosurgery. Ann R Cole Surg Eng
management of metastatic colorectal liver 1984;66:313-318.
lesions. Am Surg 2000;66:611-(j15. 25. Ishiguro H, Rubinsky B. Mechanical interactions
11. Weber SM, Lee FT, WarnerTF, Chosy SG, Mahvi between ice crystals and red-blood cells during
DM. Hepatic cryoablation: US monitoring of directional solidification. Cryobiology 1994;31:
extent of necrosis in normal pig liver. Radiology 483-500.
1998;207:73-77. 26. Kahlenberg MS, Volpe C, Klippenstein DL,
12. Steed J, Saliken JC, Donnelly BJ, Ali-Ridha NH. Penetrante RB, Petrelli NJ, Rodriguez-Bigas
Correlation between thermosensor temperature MA. Clinicopathologic effects of cryotherapy
and transrectal ultrasonography during prostate on hepatic vessels and bile ducts in a porcine
cryoablation. Can Assoc Radiol J 1997;48:186- model. Ann Surg OncoI1998;5:713-718.
190. 27. Weber SM, Lee FT, Chinn DO, Warner T, Chosy
13. Cha C, Lee FT Jr, Rikkers LF, Niederhuber JE, SG, Mahvi DM. Perivascular and intralesional
Nguyen BT, Mahvi DM. Rationale for the com- tissue necrosis after hepatic cryoablation: re-
bination of cryoablation with surgical resection sults in a porcine model. Surgery 1997;122:742-
of hepatic tumors. J Gastrointest Surg 2001;5: 747.
206-213. 28. Tatsutani K, Rubinsky B, Onik G, Dahiya R.
14. Ravikumar TS, Kane R, Cady B, Jenkins R. A Effect of thermal variables on frozen human
5-year study of cryosurgery in the treatment of primary prostatic adenocarcinoma cells. Urology
liver tumors. Arch Surg 1991;126:1520-1524. 1996;48:441-447.
15. Zhou XD, Tang ZY, Yu YQ, et al. The role of 29. Ravikumar TS, Steele G, Kane R, King V. Exper-
cryosurgery in the treatment of hepatic cancer: a imental and clinical observations on hepatic
report of 113 cases. J Cancer Res Clin Oncol cryosurgery for colorectal metastases. Cancer
1993;120:100-102. Res 1991;51:6323-6327.
16. Goering JD, Mahvi DM, Niederhuber JE, Chicks 30. Neel HB KA. Requisites for successful cryogenic
D, Rikkers LF. Cryoablation and liver resection surgery of cancer. Arch Surg 1971;102:45-48.
for noncolorectal liver metastases. Am J Surg 31. Rivoire ML, Voiglio EJ, Kaemmerlen P, et al.
2002;183:384-389. Hepatic cryosurgery precision: evaluation of
17. Onik G, Cobb C, Cohen J, Zabkar J, Porterfield ultrasonography, thermometry, and impedance-
B. US characteristics of frozen prostate. Radiol- metry in a pig model. J Surg Oncol 1996;61:
ogy 1988;168:629-631. 242-248.
18. Lee DI, McGinnis DE, Feld R, Strup SE. Retro- 32. Yeh C1. Cryosurgical treatment of melanin-
peritoneal laparoscopic cryoablation of small pigmented gingiva. Oral Surg Oral Med Oral
renal tumors: intermediate results. Urology 2003; Pathol Oral Radiol Endod 1998;86:66Q-(j63.
61:83-88. 33. Shepherd JP, Dawber RP. Wound healing and
19. Gill IS, Novick AC, Soble 11, et al. Laparoscopic scarring after cryosurgery. Cryobiology 1984;21:
renal cryoablation: initial clinical series. Urology 157-169.
1998;52:543-551. 34. Gage AA, Fazekas G, Riley EE Jr. Freezing
20. Rewcastle JC, Sandison GA, Saliken JC, injury to large blood vessels in dogs. With com-
Donnelly BJ, McKinnon JG. Considerations ments on the effect of experimental freezing of
during clinical operation of two commercially bile ducts. Surgery 1967;61:748-754.
available cryomachines. J Surg Oncol 1999;71: 35. Wong WS, Chinn DO, Chinn M, Chinn J, Tom
106-111. WL. Cryosurgery as a treatment for prostate
21. Lee FT, Chosy SG, Weber SM, Littrup PJ, Warner carcinoma: results and complications. Cancer
TF, Mahvi DM. Hepatic cryosurgery via minilap- 1997;79:963-974.
19. Cryoablation 265

36. Cox RL, Crawford ED. Complications ofcryosur- taneous cryoablation in a pig liver model: pilot
gical ablation of the prostate to treat localized study. Radiology 1999;211:687-692.
adenocarcinoma of the prostate. Urology 1995; 48. Silverman SG, Tuncali K, Adams DF, et al. MR
45:932-935. imaging-guided percutaneous cryotherapy of
37. Adam R, Akpinar E, Johann M, Kunstlinger F, liver tumors: initial experience. Radiology 2000;
Majno P, Bismuth H. Place of cryosurgery in the 217:657-664.
treatment of malignant liver tumors. Ann Surg 49. Harada J, Dohi M, Mogami T, et al. Initial expe-
1997;225:39-48. rience of percutaneous renal cryosurgery under
38. Rewcastle JC, Hahn LJ, Saliken JC, McKinnon the guidance of a horizontal open MRI system.
JG. Use of a moratorium to achieve consistent Radiat Med 2001;19:291-296.
liquid nitrogen cryoprobe performance. J Surg 50. Daniel BL, Butts K, Block WE Magnetic reso-
OncoI1997;66:11D-113. nance imaging of frozen tissues: temperature-
39. Lee F, Bahn DK, McHugh TA, Onik GM, Lee FT dependent MR signal characteristics and
Jr. US-guided percutaneous cryoablation of relevance for MR monitoring of cryosurgery.
prostate cancer. Radiology 1994;192:769-776. Magn Reson Med 1999;41:627-630.
40. Adam R, Hagopian EJ, Linhares M, et al. A com- 51. Matsumoto R, Oshio K, Jolesz FA. Monitoring
parison of percutaneous cryosurgery and percu- of laser and freezing-induced ablation in the
taneous radiofrequency for unresectable hepatic liver with Tl-weighted MR imaging. J Magn
malignancies. Arch Surg 2002;137:1332-1339. Reson Imaging 1992;2:555-562.
41. Huang A, McCall JM, Weston MD, et al. Phase I 52. Gilbert JC, Rubinsky B, Wong ST, Brennan KM,
study of percutaneous cryotherapy for colorec- Pease GR, Leung PP. Temperature determina-
tal liver metastasis. Br J Surg 2002;89:303-310. tion in the frozen region during cryosurgery of
42. Washington K, Debelak JP, Gobbell C, et al. rabbit liver using MR image analysis. Magn
Hepatic cryoablation-induced acute lung injury: Reson Imaging 1997;15:657-667.
histopathologic findings. J Surg Res 2001;95:1-7. 53. Seifert JK, Morris DL. World survey on the com-
43. Saliken JC, McKinnon JG, Gray R. CT for mon- plications of hepatic and prostate cryotherapy.
itoring cryotherapy. AIR 1996;166:853-855. World J Surg 1999;23:109-113.
44. Kaufman CS, Bachman B, Littrup PJ, et al. 54. McGahan JP, Brock JM, Tesluk H, Gu WZ,
Office-based ultrasound-guided cryoablation of Schneider P, Browning Po. Hepatic ablation
breast fibroadenomas. Am J Surg 2002;184:394- with use of radio-frequency electrocautery in the
400. animal model. J Vase Intervent Radiol 1992;3:
45. Kirchner J, Kickuth R, Laufer U, Schilling EM, 291-297.
Adams S, Liermann 0. CT fluoroscopy-assisted 55. Collyer WC, Landman J, Olweny EO, et al. Com-
puncture of thoracic and abdominal masses: a parison of renal ablation with cryotherapy, dry
randomized trial. Clin Radiol 2002;57:188-192. radiofrequency, and saline augmented radio-
46. Sandison GA, Loye MP, Rewcastle JC, et al. x- frequency in a porcine model. J Am Coil Surg
ray CT monitoring of iceball growth and thermal 2001;193:505-513.
distribution during cryosurgery. Phys Med Bioi 56. Curley SA, Izzo F, Delrio P, et al. Radio-
1998;43:3309-3324. frequency ablation of unresectable primary and
47. Lee FT Jr, Chosy SG, Littrup PJ, Warner TF, metastatic hepatic malignancies: results in 123
Kuhlman JE, Mahvi DM. CT-monitored percu- patients. Ann Surg 1999;230:1-8.
20
Combined Regional
Chemoembolization and Ablative
Therapy for Hepatic Malignancies
Michael C. Soulen and Lily Y. Kernagis

Hepatic malignancies are one of the most diffi- Combination Therapies for
cult therapeutic challenges in oncology. Cure
usually is not possible because of the predilec-
Hepatocellular Carcinoma
tion for intrahepatic recurrence, despite com- (HCC)
plete resection or ablation of the initial tumor.
Nonetheless, durable local control can be Neoadjuvant Therapy
achieved through vigilant monitoring and
aggressive multimodality therapy coordinated Resection and transplantation are effective
by a multidisciplinary team of specialists in methods to improve survival in patients with
medical, surgical, and interventional oncology. primary liver tumors. Orthotopic liver trans-
For tumors that tend not to metastasize aggres- plant (OLT) has been shown to be the best
sively beyond the liver, regional control may treatment for early-stage HCC (1–3). Neo-
benefit quality of life and overall survival. adjuvant therapies have been used to attempt
Most patients with hepatic malignancies to control tumor growth, prevent viability
have a tumor burden in excess of what can be of latent hepatic foci, induce necrosis, and
managed by percutaneous ablation techniques convert unresectable tumors to resectable
alone. While efficient in causing cell death, all ones to improve disease-free survival (DFS)
methods of thermal tissue destruction inher- and long-term overall survival (OS) (Table
ently are limited in the volume of tumor that 20.1).
can be reliably ablated. Various regional liver- Reports of neoadjuvant therapies followed
directed therapies exist that permit treatment by surgical removal of the tumor are partic-
of the entire liver volume; however, they lack ularly instructive because they provide
the efficiency of thermal ablation to cause histopathologic assessment of response to
tissue necrosis. Multimodality treatment may image-guided therapy. Preoperative transarte-
offer synergistic advantages over individual rial chemoembolization has been shown to be
methods alone. This chapter focuses on the capable of causing partial necrosis in HCC (4).
results of various combinations of liver-directed Complete necrosis occurs less commonly, but
regional and locally ablative therapies for also has been demonstrated (4–6). Oldhafer
treatment of primary and secondary hepatic et al (5) found that 29% of 21 pretransplant
malignancies. patients who received chemoembolization

266
20. Regional and Ablative Therapy for Hepatic Malignancies 267

TABLE 20.1. Neoadjuvant treatment for hepatocellular carcinoma (Hee).


Recurrence rate 3-year DFS 3-year OS
Study No. of patients (vs. control) (vs. control) (vs. control) Effective
Adachi (1993) (19) 46 52% (49%) No
Uchida (1996) (20) 60 43% (52%) 61% (71%) No
Harada (1996) (16) 105 38% (34%) 78% (68%) No
Majno (1997) (14) 49 57% (81%*) 29% (26%) 57% (47%) Yes (if downstaged or
complete necrosis)
Paye (1998) (11) 24 58% (58%) 33% (32%) 62% (66%) No
Oldhafer (1998) (5) 21 48% (54%) No
Veltri (1998) (6) 38 12% (15%) No
DiCarlo (1998) (9) 55 40% (20%*) 70% (38%*) Yes (in cirrhotic)
Lu (1999) (8) 20 55% (22%) 53% (33%*) Yes (in tumor >8cm)
Wu (1995) RCT (12) 24 78% (58%) No

RCT, randomized control trial; DFS, disease-free survival; OS, overall survival.
*Statistically significant.

developed complete necrosis, while 67% devel- who underwent hepatectomy; 44 patients
oped partial necrosis. However, no survival had chemoembolization preoperatively (20 of
benefit was noted. whom had tumor >8cm in diameter). In
The addition of percutaneous ethanol injec- patients with tumors >8cm, the relative risk of
tion (PEl) to chemoembolization for neoadju- preoperative chemoembolization for overall
vant therapy has been attempted to improve survival was 0.38 (p = .017), demonstrating that
induction of necrosis. Veltri et al (6) demon- there may be some benefit to preoperative
strated that 100% of patients who received chemoembolization in patients with stage II/III
preoperative chemoembolization and PEl tumor >8cm who undergo hepatectomy. In
had complete necrosis, while neoadjuvant another study (9), patients with tumors less
chemoembolization alone led to complete than 8 cm in diameter were shown to benefit
necrosis in 24.1 % of patients, and PEl alone led from preoperative chemoembolization. Di
to complete necrosis in 80% of patients. Tumor Carlo et al (9) found that there was a significant
recurrence occurred in patients who had improvement in 3-year OS (70% vs. 38%,
chemoembolization only and in patients who p < .02) and 3-year DFS (40% vs, 20%,p < .05)
failed to demonstrate tumor necrosis; however, in pre-liver resection patients with tumors $;5
patients selected to receive chemoemboliza- cm in diameter who received chemoemboliza-
tion alone had more advanced disease. Despite tion for HCC. A retrospective analysis per-
100% rate of complete necrosis, neoadjuvant formed by Zhang et al (10) showed that in
therapy with PEl and chemoembolization did 120 patients who had received preoperative
not significantly alter the recurrence rate or chemoembolization out of 1725 total patients
patient prognosis. Pre-OLT administration of who underwent hepatectomy for HCC,
chemoembolization was shown not to increase there was a greater DFS in patients who had
the risk of hepatic arterial complications, such received more than two chemoembolization
as thrombosis, after patients underwent OLT treatments. In contrast, a comparative study by
(7). Therefore, the role of neoadjuvant inter- Paye et al (11) demonstrated that neoadjuvant
vention in transplant candidates is limited to chemoembolization produced no significant
prevention of tumor progression beyond the decrease in tumor size or increase in DFS.
criteria for transplant eligibility. Additionally, Wu et al (12) demonstrated in
Studies that evaluate preoperative chemo- a randomized controlled trial that there was an
embolization for HCC prior to liver resection increased risk of extrahepatic recurrence and
have conflicting results. Lu et al (8) performed worse actuarial survival in patients who
a retrospective analysis of 120 cases of patients received chemoembolization prior to liver
with TNM classification stage II or III HCC resection.
268 M.e. Soulen and L.Y. Kernagis

Preoperative chemoembolization has led to for patients with initially unresectable tumors,
variable effects on preresection tumor size. In a since preoperative chemoembolization delays
study by Wu et al (12), patients who had large surgery, can complicate further chemoem-
HCCs (maximum diameter of at least 10cm), bolization, and does not provide survival
were demonstrated to be resectable by amount benefit in most studies of resectable tumors
of functional liver reserve and anatomic extent (18-20).
of tumor. Thmor volume was decreased by a
mean of 42.8% in 16 patients who received
chemoembolization. Chemoembolization did
Treatment Combinations for HCC
without Surgical Resection
not lead to complete necrosis, and there was no
change in the DFS. Additional studies on the Various nonsurgical treatment combinations
effect of neoadjuvant therapy on tumor size have been attempted for patients with HCC to
and conversion of unresectable tumors to improve prognosis. These include liver-directed
resectable have led to more favorable conclu- therapies such as radiofrequency ablation
sions (13-15). In a study by Majno et al (14), (RFA), microwave ablation, laser ablation,
improvement in DFS was demonstrated in cryoablation, chemical ablation, chemoem-
patients who were downstaged or had complete bolization, and radioembolization, as well as
necrosis, compared to patients with no response traditional systemic and radiation treatments.
or patients who did not receive preopera- Prospective randomized trials have demon-
tive chemoembolization. Additionally, 10% of strated a significant improvement in response
patients became resectable after downstaging, in those patients who receive chemoemboliza-
which occurred more frequently with tumors tion and ethanol injection compared to pa-
>5 cm. A different study (16) demonstrated no tients who receive chemoembolization alone.
significant improvement in OS and DFS; Yamakado et al (23) demonstrated that 26
however, nearly 50% of patients who under- patients with unresectable HCC who received
went neoadjuvant chemoembolization demon- chemoembolization and transportal ethanol
strated at least 25% decrease in maximum had 1,3, and 5 year survivals of 87%, 72%, and
tumor diameter (69.6% of patients with tumors 51 % (18 patients received a second round of
less than Scm). While the data on chemoem- chemoembolization). Similarly, Bartolozzi et al
bolization conversion of unresectable HCC to (24) found there was significantly greater ther-
resectable have been variable, a study using apeutic response, lower tumor recurrences, and
chemoradiation as neoadjuvant therapy by better rate of survival without recurrence in
Sitzmann (15), showed that patients with patients treated with chemoembolization
tumors that were converted to resectable after followed by PEl, compared to patients who
chemoradiation survived as long as patients received repeated chemoembolization treat-
who were resectable initially. ment for large HCCs (3.1 cm-8.0cm diameter).
Majno et al (14) showed that patients who Although chemoembolization-PEl patients
underwent preoperative chemoembolization had better survival rates (100%,87%, 72% vs.
had an increased rate of resection of adjacent 93%,70%, and 43% at 1,2, and 3 years respec-
organs (58% vs. 25%, p = .03) compared to tively), the difference was not statistically sig-
control patients. Other complications included nificant. Additionally, patients who received
adhesion of the tumor to the diaphragm, thick- chemoembolization and transportal ethanol
ening of the gallbladder wall, and thickening of injections were shown to develop more com-
the hepatoduodenalligament (17). plete tumor necrosis (25).
It appears as if preoperative chemoem- Thermal ablative techniques are limited by
bolization alone may be effective in inducing at the heat-sink effect from blood flow in the liver.
least partial necrosis and more complete necro- A few investigators have attempted to combine
sis when PEl is added to the neoadjuvant thermal ablation with alteration in hepatic blood
regimen. It has been suggested that preopera- flow to improve local tumor control. Percuta-
tive neoadjuvant therapy should be reserved neous microwave coagulation therapy was given
20. Regional and Ablative Therapy for Hepatic Malignancies 269

to 18 patients with HCC (2-3cm in diameter) Treatment for Liver Metastases


and cirrhosis within 1-2 days of chemoem-
bolization (26); 17/18 patients were demon- The liver is the most common site for colorec-
strated to have complete tumor necrosis. tal cancer recurrence. Patients with colorectal
Although one patient developed a pleural effu- cancer metastasis to the liver have a poor prog-
sion, no fatal complications presented and all nosis. Liver resection is the only potentially
patients were alive at follow-up of 12-31 months. curative option, with a 30% 5-year survival
Laser thermal ablation (LTA) was investigated (30). Patients with unresectable colorectalliver
in combination with chemoembolization in metastasis have a median survival of 6 to 9
patients with large HCCs (3.5cm-9.6cm in months without treatment (31). Multimodality
diameter) (27). LTA preceded chemoemboliza- treatment options with and without resection
tion in an attempt to make the tumor more have been sought to improve outcomes in
amenable to chemoembolization. Complete patients with liver metastasis.
tumor necrosis was seen in 90% of treated cases, Thus far, chemoembolization in combination
with cumulative survival rates of 92%, 68%, and with systemic treatment has not demonstrated
40% at 1, 2, and 3 years, respectively. Disease- promising results. In a study by Leichman et al
free survival rates were 74% and 34% at 1 and (32), chemoembolization (doxorubicin/mito-
2 years, respectively, and no major complications mycin C/cisplatin) and systemic 5-fluorouracil
resulted from laser therapy. Rossi et al (28) per- (5-FU) with leucovorin were administered to
formed percutaneous RFA in 62 patients with 31 patients with no improvement in survival
unresectable HCC (3.5 cm-8.5 cm, mean 4.7cm) compared to systemic treatment alone; the
after hepatic arterial occlusion; 90% were com- overall response was 29%, I-year survival
pletely ablated by radiologic criteria after a was 58%, and median time to progression
single RFA session, and 100% after two sessions. was 8 months. An additional study using
Local recurrence was 19% at 12 months, cisplatin and systemic 5-FU (33) yielded a
although 45% had new intrahepatic tumors. median survival of 14.3 months and overall
One-year survival was 87%, with two patients survival of 57% at 1 year and 19% at 2 years in
subsequently undergoing surgical resection. 27 patients.
The encouraging early results of combining Several combination therapies have been
thermal and embolic therapy must be tempered tested as an adjunct to resection of liver metas-
by our knowledge of the limitations of imaging tases with more favorable results. Tono et al (34)
in measuring response. Unpublished data from demonstrated a beneficial effect of prophylac-
our institution are sobering-90% of hepatomas tic hepatic arterial infusion of 5-FU after resec-
treated with one or more image-guided proce- tion of colorectal metastases in nine patients (10
dures prior to liver transplantation harbor controls). Median DFS was 78%,78%, and 67%
viable tumor at the time of explantation, at 1,2, and 3 years, respectively, for the treated
despite "complete" radiologic responses. Fur- group versus 50%,30%, and 20%. Additionally,
thermore, the lack of controlled studies that Kemeny et al (35) studied adjuvant hepatic
evaluate the impact of local treatment on the arterial infusion of chemotherapy (fluoxuri-
natural history of the disease is an important dine/dexamethasone) through an implantable
challenge for percutaneous therapy, particu- pump after resection of hepatic metastases from
larly given the high prevalence of new tumors colorectal cancer in 74 patients. There was sig-
that appear in the liver. A nonrandomized, nificant improvement in 2-year OS compared to
uncontrolled single-institution comparison of the untreated group (86% vs. 72%) and hepatic
37 patients undergoing either chemoemboliza- progression-free survival at 2 years (90% vs.
tion alone (n = 24) or chemoembolization plus 60%). The benefit seemed to be confined to
RFA (n = 13) showed no difference in objective patients not treated previously with systemic
response, but a significant improvement in chemotherapy.
mean and I-year survival with the combined A randomized control trial by Lorenz et al
therapy (29). (36) demonstrated no significant improvement
270 M.e. Soulen and L.Y. Kernagis

in 226 patients with colorectal cancer metas- 3. Selby R, Kadry Z, Carr B, Tzakis A, Madariaga
tases to the liver who underwent hepatic artery JR, Iwatsuki S. Liver transplantation for hepa-
infusion following curative resection. Patients tocellular carcinoma. World J Surg 1995;19(1):
had a median survival of 34.5 months vs. 46.8 53-58.
months in the control group, and time to pro- 4. Spreafico C, Marchiano A, Regalia E, et a1.
Chemoembolization of hepatocellular carci-
gression of 14.2 months vs. 13.7 months.
noma in patients who undergo liver transplanta-
Rose et al (37) evaluated RFA in patients tion. Radiology 1994;192:687-690.
with hepatic metastases as a primary treatment 5. Oldhafer KJ, Chavan A, Fruhauf NR, et a1. Arte-
or as an adjunct to resection, cryosurgical abla- rial chemoembolization before liver transplanta-
tion, or hepatic artery infusion. They demon- tion in patients with hepatocellular carcinoma:
strated no RFA-associated deaths, and only marked tumor necrosis, but no survival benefit?
minimal local and systemic complications. This J Hepatol 1998;29:953-959.
was confirmed in a study by Elias et al (38) in 6. Veltri A, Grosso M, Martina MC, et a1. Effect of
patients who underwent RFA for a central preoperative radiological treatment of hepato-
metastasis or as an adjunct to hepatectomy in cellular carcinoma before liver transplantation: a
patients with multiple bilateral metastases; retrospective study. Cardiovasc Intervent Radiol
1998;21:393-398.
RFA increased the rate of curative resection
7. Richard HM III, Silberzweig JE, Mitty HA,
without complications related to the RFA. Lou WYW, Ahn J, Cooper JM. Hepatic arterial
Further studies would be needed to evaluate complications in liver transplant recipients
effect on survival and recurrence. treated with pretransplantation chemoemboliza-
tion for hepatocellular carcinoma. Radiology
2000;214:775-779.
8. Lu C-D, Peng S-Y, Jiang X-Co Chiba Y, Tanigawa
Summary N. Preoperative transcatheter arterial chemoem-
bolization and prognosis of patients with
The role of neoadjuvant and adjuvant liver- hepatocellular carcinomas: retrospective analy-
directed therapies in patients undergoing resec- sis of 120 cases. World J Surg 1999;23:293-
tion of liver tumors remains controversial. For 300.
the majority of patients who have unresectable 9. Di Carlo V, Ferrari G, Castoldi R, et a1. Preoper-
disease, an impressive armamentarium of tech- ative chemoembolization of hepatocellular
niques is available for image-guided treatment. carcinoma in cirrhotic patients. Hepato-
Combinations of these therapies may improve Gastroenterology 1998;45:1950-1954.
10. Zhang Z, Liu Q, He J, Yang J, Yang G, Wu M. The
the efficiency of tumor cell kill and result in
effect of preoperative transcatheter hepatic arte-
better immediate local control. Whether this
rial chemoembolization on disease-free survival
improvement in local response alters the after hepatectomy for hepatocellular carcinoma.
natural history of the disease remains to be Cancer 2000;89:2606-2612.
evaluated in carefully designed and controlled 11. Paye F, Jagot P, Vilgrain V, Farges 0, Borie D,
trials. Belghiti 1. Preoperative chemoembolization of
hepatocellular carcinoma. Arch Surg 1998;133:
767-772.
References 12. Wu C-C, Ho Y-Z, Ho WL, Wu T-C, Liu T-J,
P'eng F-K. Preoperative transcatheter arterial
1. Mazzaferro V, Regalia E, Doci R, et a1. Liver chemoembolization for resectable large hepato-
transplantation for the treatment of small hepa- cellular carcinoma: a reappraisal. Br J Surg 1995;
tocellular carcinomas in patients with cirrhosis. 85:122-126.
N Engl J Med 1996;334(11):693-699. 13. Tang Z-Y, Yu Y-Q, Zhou X-D, et a1. Cytoreduc-
2. Pichlmayr R, Weimann A, Oldhafer KJ, et a1. tion and sequential resection for surgically
Role of liver transplantation for the treatment verified unresectable hepatocellular carcinoma:
of unresectable liver cancer. World J Surg 1995; evaluation with analysis of 72 patients. World J
19(6):807-813. Surg 1995;19:784--789.
20. Regional and Ablative Therapy for Hepatic Malignancies 271

14. Majno PE, Adam R, Bismuth H, et al. Influence of transcatheter arterial chemoembolization and
of preoperative transarteriallipiodol chemoem- transportal ethanol injection. Radiology 1994;
bolization on resection and transplantation for 193:75-80.
hepatocellular carcinoma in patients with cir- 26. Seki T, Tarnai T, Nakagawa T, et al. Combination
rhosis. Ann Surg 1997;226(6):688-703. therapy with transcatheter arterial chemoem-
15. Sitzmann 1. Conversion of unresectable liver bolization and percutaneous microwave coagu-
cancer: an approach and follow-up study. World lation therapy for hepatocellular carcinoma.
J Surg 1995;19:790-794. Cancer 2000;89(6):1245-1251.
16. Harada T, Matsuo K, Inoue T, Tamesue S, Inoue 27. Pacella C, Bizzarri G, Cecconi P, et al. Hepato-
T, Nakamura H. Is preoperative hepatic arterial cellular carcinoma: Long-term results of com-
chemoembolization safe and effective for hepa- bined treatment with laser thermal ablation and
tocellular carcinoma? Ann Surg 1996;224(1):4-9. transcatheter arterial chemoembolization. Radi-
17. Yu Y-Q, Xu D-B, Zhou X-D, Lu J-Z, Tang Z-Y, ology 2001;219:669-678.
Mack P. Experience with liver resection after 28. Rossi S, Garbagnati F, Lencioni R, et al. Percu-
hepatic arterial chemoembolization for hepato- taneous radio-frequency thermal ablation of
cellular carcinoma. Cancer 1993;71:62-65. nonresectable hepatocellular carcinoma after
18. Poon R, Fan S-T, Wong 1. Risk factors, pre- occlusion of tumor blood supply. Radiology
vention, and management of postoperative 2000;217:119-126.
recurrence after resection of hepatocellular 29. Bloomston M, Binitie 0, Fraiji E, et al. Trans-
carcinoma. Ann Surg 2000;232(1):10-24. catheter arterial chemoembolization with or
19. Adachi E, Matsumata T, Nishizaki T, Hashimoto without radiofrequency ablation. I: The manage-
H, Tsuneyoshi M, Sugimachi K. Effects of pre- ment of patients with advanced hepatic malig-
operative transcatheter arterial chemoem- nancy. Am Surg 2002;68:827-831.
bolization for hepatocellular carcinoma: the 30. Registry of Hepatic Metastases. Resection of
relationship between postoperative course and the liver for colorectal carcinoma metastases: a
tumor necrosis. Cancer 1993;72:3593-3598. multi-institutional study of indications for resec-
20. Uchida M, Kohno H, Kubota H, et al. Role tion. Surgery 1988;103(3):278-288.
of preoperative transcatheter arterial oily 31. Wagner JS, Adson MA, Van Heerden JA, Adson
chemoembolization for resectable hepatocellu- MH, Ilstrup DM. The natural history of hepatic
lar carcinoma. World J Surg 1996;20:326-331. metastases from colorectal cancer: a comparison
21. Tanaka K, Nakamura S, Numata K, et al. Hepa- with resective treatment. Ann Surg 1984;199(5):
tocellular carcinoma: Treatment with percuta- 502-508.
neous ethanol injection and transcatheter 32. Leichman CG, Jacobson JR, Modiano M, et al.
arterial embolization. Radiology 1992;185:457- Hepatic chemoembolization combined with
460. systemic infusion of 5-fluorouracil and bolus
22. Matsui 0, Kadoya M, Yoshikawa J, et al. Small leucovorin for patients with metastatic colorec-
hepatocellular carcinoma: treatment with sub- tal carcinoma. Cancer 1999;86(5):775-781.
segmental transcatheter arterial embolization. 33. Bavisotto LM, Patel NH, Althaus SJ, et al.
Radiology 1993;188:79-83. Hepatic transcatheter arterial chemoemboliza-
23. Yamakado K, Nakatsuka A, Tanaka N, tion alternating with systemic protracted contin-
Matsumura K, Takase K, Takeda K. Long-term uous infusion 5-fluorouracil for gastrointestinal
follow-up arterial chemoembolization combined malignancies metastatic to liver: a phase II trial
with transportal ethanol injection used to treat of the Puget Sound Consortium (PSOC 1104).
hepatocellular carcinoma. JVIR 1999;10:641- Clin Cancer Res 1999;5(1):95-109.
647. 34. Tono T, Hasuike Y, Ohzato H, Takatsuka Y,
24. Bartolozzi C, Lencioni R, Caramella D, et al. Kikkawa N. Limited but definite efficacy of pro-
Treatment of large HCC: Transcatheter arterial phylactic hepatic arterial infusion chemotherapy
chemoembolization combined with percuta- after curative resection of colorectalliver metas-
neous ethanol injection versus repeated tran- tases. Cancer 2000;88(7):1549-1556.
scatheter arterial chemoembolization. Radiology 35. Kemeny N, Huang Y, Cohen AM, et al. Hepatic
1995;197:812-818. arterial infusion of chemotherapy after resection
25. Yamakado K, Hirano T, Kato N, et al. Hepato- of hepatic metastases from colorectal cancer.
cellular carcinoma: treament with a combination N Engl J Med 1999;341(27):2039-2048.
272 M.e. Soulen and L.Y. Kernagis

36. Lorenz M, Muller H-H, Schramm H, et al. Ran- primary and adjunctive ablative technique for
domized trial of surgery versus surgery followed hepatic malignancies. Am Surg 1999;65:1009-
by adjuvant hepatic arterial infusion with 5- 1014.
fluorouracil and folinic acid for liver metastases 38. Elias D, Debaere T, Muttillo I, Cavalcanti A,
of colorectal cancer. Ann Surg 1998;228(6):756- Coyle C, Roche A. Intraoperative use of radio-
762. frequency treatment allows an increase in the
37. Rose DM, Allegra DP, Bostick PJ, Foshag U, rate of curative resection. J Surg OncoI1998;67:
Bilchik Al Radiofrequency ablation: a novel 190-191.
21
Focused Ultrasound for
Tumor Ablation
Clare Tempany, Nathan MacDonold, Elizabeth A. Stewart,
and Kullervo Hynynen

This chapter reviews the basic principles of this chapter may be found in several review
focused ultrasound therapy. The physical prin- articles (4,7-10).
ciples are discussed and explained. The techni-
cal requirements and instrumentation used are
illustrated. The critical value of using magnetic Propagation Through Tissue
resonance (MR) as an image guidance method
for planning, delivering, and monitoring this Ultrasound is a form of vibrational energy (at
form of ablation therapy is reviewed, and a frequency above 20,000Hz) that is propa-
its application in uterine leiomyomas is gated as a mechanical wave by the motion of
highlighted as an example of current clinical molecules within tissue. The propagating pres-
practice. sure wave causes the tissue molecules to form
The only known method to cause highly compressions and rarefactions, such that the
localized tissue destruction deep in the body molecules oscillate around their rest position.
noninvasively is ultrasound. The use of ultra- For medical purposes, the ultrasound frequency
sound for tissue destruction was demonstrated is typically between 0.5 and 10 MHz. The speed
first in the 1940s. An extensive research period of the propagating wave is independent of the
followed and led to clinical tests of focused frequency, and is about 1500 mls in soft tissue
ultrasound surgery in the treatment of brain and water. Thus, the wavelength in soft tissue at
disorders (1,2). These early experiments, 1MHz is about 1.5 mm, and at 2 MHz it is about
however, did not lead to the use of ultrasound 0.75mm.
in routine practice due to the lack of image Ultrasound can be generated by piezoelectric
guidance and the complexity of the procedure. transducers that are driven by a radiofrequency
The recent success of minimally invasive (RF) voltage at the frequency of the desired
surgeries and the ability to combine this sound wave. In each piezoelectric element, the
technology with online image guidance and thickness changes proportionally to the ampli-
monitoring has led to a renewed interest in tude of the applied voltage, so the electrical
focused ultrasound ablation therapy. signal is converted to mechanical motion of
The first commercial clinical devices utilized the same frequency. The generated acoustic
diagnostic ultrasound to aim the therapeutic power is directly proportional to the driving
beam to the target volume. These devices have RF power. The piezoelectric elements are
been tested in the treatment of eye (3), prostate most often made of a ceramic polycrystal that
(4,5), bladder, and kidney (6) disorders with can be manufactured into the desired shape,
encouraging results. More detailed descriptions such as a flat plate to generate planar waves, or
of the history of focused ultrasound surgery a spherically curved bowl to generate focused
with references to the studies not covered in fields.

273
274 C. Tempany et al

As the ultrasound energy passes through


tissue, it is attenuated according to an expo-
nential law. At 1MHz, the ultrasound power
is attenuated approximately 50% while it prop-
agates through 7 em of tissue. At 2MHz the
wave is reduced to approximately 25 % of its
initial value by the same tissue. The attenuated
energy is converted into thermal energy in the
tissue.
Ultrasound is effectively transmitted from
one soft tissue layer to another, with a small
amount (a few percent) of the wave reflected
back. At soft tissue-bone interfaces, about one
A
third of the incident energy is reflected back at
normal incidence. In addition, the amplitude
~
attenuation coefficient of ultrasound is about ~0.8
10 to 20 times higher in bone than in soft ~
~0.6
tissue. This causes the transmitted beam to be
~
absorbed rapidly, resulting in a hot spot at the ~ 0.4
bone surface (11,12). At a soft tissue-gas inter- o
zO.2
face, all of the energy is reflected back.
00·········20········40 60' '80' 100·······
Depth (mm) B
Focused Beams FIGURE 21.1. A focused ultrasound field can be pro-
duced by a spherically curved transducer (A). The
Similar to optics, ultrasonic beams can be induced temperature rise in the targeted tissue is
focused by radiators, lenses, or reflectors. The directly proportional to the ultrasound intensity,
wavelength limits the size of the focal region which is highly localized in the focal region (B). This
(13). A focal diameter of 1 mm can be achieved localization ensures that tissue outside of the focal
in practice at 1.5 MHz with a sharply focused zone is not damaged. Acoustic lenses, reflectors, and
transducer (F number <1). The length of the arrays of small transducers ("phased arrays") also
focus is typically five to 20 times larger than the can be used to create a focused ultrasound field.
diameter. Since the ultrasound beam is trans- Phased arrays also can be utilized to move the focus
mitted from an applicator that is large in diam- and create patterns of multiple foci.
eter compared to the focal spot diameter, the
ultrasound intensity at the focal spot can be
several hundred times higher than in the over-
lying tissue. Similarly, the ultrasound exposure
Tissue Effects
drops off rapidly across the focus, thus limiting The ultrasound beam can interact with tissue
the high ultrasound exposure to the focus by elevating the temperature due to energy
(Fig. 21.1). absorption from the wave. The temperature ele-
Similar focusing can be achieved by using vation during short (a few seconds) sonications
transducer arrays that are driven with signals follows closely the ultrasound field distribution.
that have the proper phase difference to obtain At longer exposures, thermal conduction and
a common focal point (electrical focusing). perfusion smooth the temperature distribution
Arrays with a large number of elements allow and result in less sharp gradients and a more
multiple focal points to be induced simultane- variable temperature elevation. The induced
ously. Large tissue volumes thus can be exposed temperature elevation is directly proportional
with such arrays with optimized energy deposi- to the rate of energy absorption, but it also
tion patterns (12,14). depends on the heat transfer by thermal con-
21. Focused Ultrasound for Tumor Ablation 275

duction and blood perfusion in the tissue. In Lynn et al (26) investigated the potential sur-
addition, tissue close to large blood vessels will gical application of focused ultrasound (FUS)
be cooled by the flow (15-17). Since the ultra- over six decades ago. Since then, therapeutic
sound absorption coefficient and the blood ultrasound has been tested extensively for
perfusion are highly variable from location to noninvasive surgery both in animals and
location, even in the same organ, the absolute humans (27-29). During the past two decades,
temperature elevation is difficult to predict clinical trials using focused ultrasound for
based on the power output of the transducer. noninvasive surgery of the prostate and other
Therefore, temperature monitoring is required sites have been conducted with diagnostic
if predictable temperature elevations are to be ultrasound for localization and targeting (30).
induced. Although the use of FUS for ablation of malig-
Different degrees of thermal damage to the nant tumors has been shown to be promising
tissue depend on the temperature reached and (31,32), its widespread acceptance has been
the duration of the exposure (18-21). For expo- limited because of the lack of precise target def-
sures of a few seconds, temperatures of above inition and the difficulty in controlling the focal
approximately 60°C are needed to cause irre- spot position and beam dosimetry without a
versible tissue damage (24). The elevated tem- temperature-sensitive imaging method.
perature can block the microvasculature and Magnetic resonance imaging (MRI) can
stop blood perfusion in the coagulated tissue satisfy these requirements for FUS therapy
volume. Occlusion of larger blood vessels by (33-37). It has excellent anatomic resolution for
thermal effects of ultrasound also has been targeting, high sensitivity for localizing tumors,
demonstrated (22-24). The temperature eleva- and temperature sensitivity for online treat-
tion also may be useful to localize gene therapy ment monitoring. Several MRI parameters are
(25). When the temperatures reach lOO°C, the temperature sensitive; the one based on the
tissue water boils and gas formation results. The proton resonance frequency allows relatively
gas blocks the propagation of the ultrasound small temperature elevations to be detected
beam and significantly modifies the energy prior to any irreversible tissue damage (38).
deposition pattern. Thus, the location of the focus can be detected
at relatively low powers, and the accuracy of
targeting can be verified. In addition, using cal-
Advantages and Disadvantages of ibrated temperature-sensitive MRI sequences,
Focused Ultrasound focal temperature elevations and effective
The main advantages of using focused ultra- thermal doses may be estimated (39,40). Such
sound are that it is noninvasive and that it can thermal quantificaton allows for online feed-
be focused. This allows deep targets to be back to ensure that the treatment is safe, by
destroyed without any damage to the overlying assuring that the focal heating is confined to the
or surrounding tissue. These characteristics also target volume and below the level for boiling.
make it possible to deliver the energy rapidly, Thermal assessment predicts effectiveness by
thus minimizing the effects of perfusion and assuring that the temperature history is suffi-
thermal conduction; these factors result in cient to ensure thermal coagulation.
sharp temperature gradients and accurate The technical feasibility of performing MRI-
tissue coagulation. guided focused ultrasound surgery utilizing
The main disadvantages of focused ultra- MRI both to guide and monitor the therapy
sound are its strong attenuation in bone, and has been established in animal experiments
its reflection at gas interfaces, which limits (40-42). Clinical feasibility has been demon-
the use of ultrasound to sites with a soft tissue strated on benign and malignant tumors of the
window for beam propagation. In addition, breast (43-45). Adequacy of treatment of target
the sharp focusing of ultrasound makes the volumes can be substantiated not only by
treatment time long when large tumors are intraprocedural, temperature-sensitive MRI,
treated. but also by using postprocedural T1- and T2-
276 C. Tempany et al

weighted imaging, which reveals signal intensity or on the peritoneal surface in a diffuse manner
changes in the treated tissue (46-48). Occlu- know as leiomyomatosis.
sion of the microvasculature within sonicated Uterine leiomyomas do not always require
tissue can be detected by posttreatment MRI treatment. Although many authors recommend
with intravenous contrast agent administration treatment with drugs such as birth control pills,
(47,49). there is little evidence to support this practice
(61). Thus, the bulk of treatment options for
fibroids is surgical.
Uterine Leiomyomas Numerous factors influence treatment
options, including the size, number, and loca-
Leiomyomas are very common and cause a tion(s) of the myomas, the presenting symp-
range of clinical symptoms, from severe bleed- toms, age, and reproductive desires of the
ing to minor discomfort (50). Treatment options woman.
include hysterectomy, myomectomy, uterine
artery embolization, and hormonal therapy.
Thermal ablation of uterine leiomyomas by
Surgical Therapies
percutaneous interstitial laser therapy or Abdominal myomectomy has been the tradi-
cryoablation has been investigated as a poten- tional alternative to hysterectomy, since it pre-
tial minimally invasive treatment (51-53). serves the uterus and allows childbearing (62).
Focused ultrasound, which delivers the thermal However, it does have morbidity, similar to that
energy without the need to insert a probe, has of hysterectomy, and it entails the significant
the potential to become a fully noninvasive possibility that subsequent surgery may be
choice for selected patients. We designed a needed (63-65). For some fibroids, an endo-
study to test the feasibility and safety of MRI- scopic approach is possible with either laparo-
guided FUS for treatment of benign leiomy- scopic or hysteroscopic myomectomy (55).
omas of the uterus. However, the size, location, and number of
Uterine leiomyomas, or myomas or fibroids, fibroids eliminate this option for many women.
are the most common pelvic neoplasm in Uterine artery embolization (UAE) is
women; they occur in 20% to 50% of women increasingly the first-line alternative to hys-
over 30 years of age (50). Their clinical impor- terectomy for women with bulk-related symp-
tance is that they are the leading indication for toms and no desire for future pregnancy. It
hysterectomy (54). They are treated by less decreases menorrhagia and bulk-related symp-
invasive surgical options including myomec- toms in most women (66,67). Serious complica-
tomy and uterine artery embolization (55,56). tions are rare, but appear to be increased when
Fibroids account for significant morbidity in there is a single large myoma (68-71).
terms of both pelvic pressure and heavy men- There also appears to be an age-related risk
strual bleeding (57). of ovarian failure that limits its use for women
The incidence of fibroids is increased in black desiring fertility (67).
women (58,59). This population of women
develops more advanced disease by the time
of surgical intervention, and has surgery at a
Magnetic Resonance Imaging of
younger age (60).
Uterine Leiomyomas
Pathologically, fibroids are smooth muscle One of the most important roles of MRI in the
tumors composed of whorls of muscle fibers body is imaging the female pelvis, as MRI has
arranged in circular fashion. They are well- well-defined and established roles in imaging
defined focal masses that occur either intramu- many diseases of the female pelvis. These roles
rally in the myometrium, or are submucosal, range from evaluating congenital anomalies to
subserosal, or pedunculated from the uterine staging gynecologic neoplasms, such as cervical
surface. Rarely, fibroids arise in extrauterine cancer. One of the earliest applications was in
locations such as the broad ligament or ovary, the definition of pelvic masses that could not be
21. Focused Ultrasound for Tumor Ablation 277

diagnosed and characterized by ultrasound. locations of the bladder, bowel, and spine are
Magnetic reSOnance imaging of the pelvis is important, as will be detailed in the procedure
performed, irrespective of magnet field description below. To classify myomas by
strength, using a combination of Tl- and T2- their symptoms, it is important to define their
weighted sequences. The female genital organs location, for example, submucosal Ones are
vary in position and orientation, and thus more likely to cause bleeding and anemia,
multiplanar imaging is essential. while subserosal myomas typically have "bulk"
Leiomyomas vary both pathologically and on symptoms, such as pressure On the bladder.
imaging. They vary in size, location, internal Although it is difficult to assign specific symp-
tissue character, and behavior. Their wide toms to individual myomas, it is important to
variety in appearance can explain, in part, their document the details of each prior to focal
varied symptoms. On imaging they are classi- therapy.
cally large well-defined masses in the uterus. In
many situations, a bulky uterus is often classi-
fied as "fibroid". Magnetic resonance provides SizeNolume
the most accurate imaging of myomas cur-
Magnetic reSOnance imaging obtains the truest
rently. It can provide a full detailed assessment
measurement of in vivo size and volume. Ultra-
of each and every myoma in the uterus. This
sound can provide the accurate size of the
typically includes the number, sizes, and loca-
uterus and estimates of individual myomas. For
tions of all myomas in the uterus. However,
treatment monitoring, three-dimensional (3D)
there are many other features that require
MR volumetric assessment before and after
more attention and detailed examination. As
therapy is a useful way to assess response.
focal ablative treatments for leiomyomas are
investigated, the baseline imaging characteris-
tics must be well understood, before interven-
ing for new treatment. These focal therapies
Tissue Characteristics
include uterine artery embolization, laser, and Magnetic resonance imaging can demonstrate
focused ultrasound surgery. six patterns in uterine leiomyomas:
1. Classic
Location 2. Hypercellular
3. Cystic degeneration
Location of leiomyomas can be assessed in
4. Hemorrhagic/carneous or "red" degenera-
three dimensions, which is critical for treatment
tion
planning. Uterine artery embolization is not felt
5. Necrotic
to be effective for large pedunculated myomas.
6. Lipoleiomyomas
For planning FUS, it is essential to define the
location of the myoma, and most importantly The typical signal intensity pattern of each of
its relationship to surrounding tissue. The these is listed in Table 21.1. It is important to

TABLE 21.1. Typical signal intensity patterns of uterine leiomyomas.


Type Tl-weighted signal T2-weighted signal Postgadolinium
Classical Isointense Low signal Homogeneous enhancement
Hypercellular Isointense High signal Homogeneous enhancement
Cystic degeneration Iso-low signal Hyperintense areas with No enhancement in hyperintense areas
surrounding low signal
Carneous Hyperintense Mixed, low signal Nonenhancing
degeneration
Necrotic Low signal Hyperintense Nonenhancing
Lipoleiomyomas High signal High signal Nonenhancing
278 C. Tempany et al

classifiy the myoma type prior to treatment


because it is unlikely that an ablative treatment
or embolization will be effective on necrotic
or degenerated myomas. The effects of these
therapies based on myoma type is not well
understood.

Magnetic Resonance-Guided
Focused Ultrasound Surgery for
Uterine Leiomyomas
This procedure is being evaluated in the stan-
dard fashion with phase I, II, and III trials. Cur- FIGURE 21.2. Patient in a magnetic resonance imager
rently, we are in the latter stages of a phase III lying on the focused ultrasound (FUS) system table.
trial. The feasibility and safety of this method
were assessed in multicenter, multinational
phase I and II trials. In these trials, all women
who were enrolled were scheduled for hysterec- position, it was important to evaluate the
tomy for their symptomatic uterine leiomy- images for possible obstacles to treatment such
omas. They agreed to undergo FUS treatment 1 as bowel loops between the leiomyoma and the
week prior to surgery. anterior abdominal wall. MRI images were
The eligibility criteria for treatment in the used to determine which leiomyomas could be
protocol included: (1) patient was eligible for treated safely. If there were multiple leiomy-
MRI, and had no pacemaker, claustrophobia, or omas in safe locations, the largest one was
other contraindication to MRI; (2) patient had selected. The goal of this phase of the trial was
no midline abdominopelvic scars; (3) the size of to deliver a treatment that could safely induce
the uterus was 20cm or less; (4) the size of the thermocoagulation that was demonstrable
myomas was lOcm or less; (5) patient had no pathologically.
intervening bowel or bladder between the All MRI examinations were performed on a
transducer and the myoma. I.5-T standard whole-body system (Signa, GE
Medical System) with the patient lying prone
(Fig. 21.2). Standard T2-weighted fast spin echo
Treatment Planning
(FSE) images were obtained in three planes
All patients underwent screening in the clinic, through the uterus, using either a body coil or
and questionnaires were administered to docu- an external multicoil array. Initial localizer
ment symptoms. All women had pretreatment large field-of-view T2-weighted images were
MRI scans using a standardized protocol that acquired to localize the transducer, uterus, and
included Tl- and T2-weighted images before leiomyomas (Fig. 21.3). Typical parameters
and after administration of intravenous (IV) used for the T2-weighted FSE sequence were
gadolinium. The MR images defined the field of view (FOV) 160 to 250mm, matrix size
leiomyomas by size, volume, and location, and 256 x 192, TR 4 to 5000ms, TE 90 to 120ms,
demonstrated enhancement after IV gadolin- three data acquisitions, slice thickness 4mm,
ium contrast. Magnetic resonance imaging also with a I-mm gap, BW = 16Hz. Then Tl-
was used to plan the beam path by ensuring weighted SE sequences were obtained: FOV
that each targeted leiomyoma was in an acces- 160-250, matrix size 256 x 128, TRITE = 600120
sible location. ms, four data acquisitions, slice thickness
As FUS is delivered through the anterior 4/1 mm. For contrast-enhanced images, Tl-
abdominal wall with the patient in the prone weighted spoiled gradient recalled echo
21. Focused Ultrasound for Tumor Ablation 279

A B

FIGURE 21.3. (A-C) Magnetic resonance imaging scans


of leiomyoma. MuItiplanarT2-weighted images show the
typical low signal intensity appearance of a large intra-
mural leiomyoma and small submucosal leiomyoma
(arrows). Scans are axial, sagittal, and coronal. c

(SPGR) imaging started very soon after IV perfusion of the myomas and the uterus, and (2)
injection of the gadolinium-based contrast to provide information regarding location of
agent (dose 0.1 mmollkg body weight, Mag- the leiomyoma and its relationship to adjacent
nevist, Berlex Laboratories, Wayne, NJ). Pre- structures. All patients were imaged in the
treatment and posttreatment follow-up MRIs prone treatment position and on the treatment
were obtained according to the same protocol. table containing the FUS transducer (Fig.
Gadolinium serves two purposes: (1) to assess 21.4A-C).
280 C. Tempany et al

A-C

FIGURE 21.4. Atypical hypercellular leiomyoma on during two focused ultrasound exposures. Multiple
MR. (A) Woman with history of prior cervical cancer ultrasound exposures at overlapping locations were
presents with large pelvic mass. Sagittal TI-weighted delivered to treat a volume in the fibroid. These maps
image shows large, well-defined high signal intensity were used to guide the treatment; they ensured that
mass in the myometrium. (B) Sagittal pregadolinium the ultrasound was focused in the correct location,
Tl-weighted fat-saturated image shows homoge- and they verified that the heating was sufficient.
neously low signal intensity mass. (C) After the bolus Right: Contrast-enhanced images acquired after the
IV injection of 20cc of gadolinium, the mass shows treatment showing the treated area, which appears
heterogeneous enhancement. (D) Left: T2-weighted as a dark region. The image in the top row was
(top left) and contrast-enhanced Tl-weighted acquired in a sagittal plane, which is parallel to the
(bottom left) MR images acquired before the treat- direction of the ultrasound beam. The image in the
ment. The contrast-enhanced image shows that the bottom was acquired in the coronal plane, and is
entire fibroid was perfused before the ultrasound perpendicular to the direction of the ultrasound
treatment. Center: Temperature maps acquired beam.
21. Focused Ultrasound for Tumor Ablation 281

MRg Focused Ultrasound rounding the transducers and providing skin


Equipment coupling. Pretreatment imaging is performed
with T2-weighted FSE images in three orthog-
All treatments have been performed using an onal planes. The radiologist outlines the volume
MRg FUS system (ExAblate 2000, InSightec- to be treated in the leiomyoma, based on
TxSonics, Haifa, Israel). A focused, piezoelec- selected slices from the coronal T2-weighted
tric transducer with a 120-mm diameter and images, using the system software. The target is
operating frequency of 1.0 to 1.5 MHz gener- visualized in two imaging planes, usually
ates the ultrasound field. It is located in the MR coronal and sagittal. The computer software in
table, surrounded by a water bath. It can elec- the system allows display of the ultrasound
tronically control the location of the focal spot beam overlying all tissues through which it
and the volume of the coagulated tissue. A thin will pass. This permits evaluation of all
plastic membrane window covers the water tissues or structures in the beam path (Fig.
tank and allows the ultrasound to propagate 21.4D).
into the patient's pelvis. All elements, cables, The T2-weighted images allow assessment of
parts, and components have been manufac- the beam path and determine if a safe treat-
tured to be MR compatible and function in an ment can be delivered. Care is taken to avoid
MR environment. The system operates inside a contact with bowel loops, and there must be
1ST magnet (GE Signa, General Electric no bowel in the beam path. If necessary, the
Medical Systems, Milwaukee, WI). beam can be repositioned or even tilted to
optimize the path. The entire beam path must
be assessed, including the posterior path. It
Patient Preparation is best to avoid the beam directly passing
The night before the procedure the patient through the spine, as bone absorbs a significant
shaves all hair from the pubic bone upward to amount of the energy and can cause local bone
allow optimal skin coupling with the treatment pain.
transducer. The patient gets an IV for conscious After an appropriate volume is selected, the
sedation to reduce pain and prevent motion. sonication plan is developed in the computer.
This is achieved using either oral antianxiolyt- This initial plan places equally spaced over-
ics, such as diazepam, or conscious sedation lapping focal volumes such that the entire
with IV fentanyl and Versed. Patients who target can be covered. The positioning of the
receive conscious sedation are monitored focal sonications is selected so that the induced
during the procedure by a nurse, with routine tissue coagulation induces complete coverage
heart rate, blood pressure, and oxygen satura- in the selected volume. The volume to be
tion level measurements. The decision to use treated is calculated by measuring the volume
concious sedaton is based on the patient's level of the sonication cylinder, which is based on the
of comfort when lying prone during the initial size of the focal spot; the latter ranges from 4.5
MRI, and on whether she has complaints of to 6.0mm, and the spot length ranges from 18.0
joint pain, such as shoulder or neck pain. to 28.0mm.
During the procedure the patient is asked to The locations of the planned sonications
report any symptoms, especially pain or heat. can be modified during the treatment by the
These are reported to the nurse in the room and operator to obtain accurate MRI thermometry-
the radiologist at the treatment console outside derived thermal dose and complete coverage of
the room. the target volume. In each treatment, all of the
sonications are at the same depth (i.e., only one
plane is sonicated). The T2-weighted images are
used to outline and define the actual target for
Pretreatment Imaging therapy manually in the treatment planning
The patient is positioned prone in the magnet software. The total number of sonications is
on the transducer with the water bath sur- calculated and the plan confirmed.
282 C. Tempany et al

Treatment treatment. To this end, a second study follows


women with symptomatic fibroids before and
Before the therapy-level sonications, low-
after treatment, using a quality of life tool to
energy test pulses are aimed within the target
measure treatment effect.
volume. The power is increased, until the loca-
tion of the focus is visible on the temperature-
sensitive images. These sonications are below
Conclusion
the level for thermal tissue damage, and rather
Clinical trials using FUS or MR-guided FUS
are used to align the coordinates of the FUS
are under way in many sites, with various clin-
system with the MR coordinates. When the test
ical applications. Focused ultrasound of uterine
pulse is located in the planned position, the
fibroid tumors is feasible and safe in initial
complete target volume is sonicated with a
trials. This form of noninvasive treatment has
series of higher power pulses such that ade-
the potential to become a major ablative
quate temperature and thermal dose are
method that may well have broad appeal to
reached. The pulse duration typically is 16
patients and their physicians.
seconds. The interval between pulses is about 3
minutes. This allows for cooling of the tissues
between each thermal treatment. During each
References
treatment the number of sonications delivered 1. Fry WJ, Barnard JW, Fry EI Ultrasonic lesions
is recorded, as is the number of visible MR in the mammalian central nervous system.
temperature maps, the power used, and the Science 1955;122:517-518.
temperature changes. 2. Fry WJ, Fry FI Fundamental neurological
research and human neurosurgery using intense
Thus, initial sonications are delivered at low
ultrasound. IRE Trans Med Electron 1960;ME-
power, followed by slow power increases to 7:166-181.
ensure that the patient is comfortable and that 3. Coleman D, Lizzi FL, Driller I Therapeutic
the thermal imaging demonstrates precise ultrasound in the treatment of glaucoma.
treatment location. Using phase maps, each Ophthalmology 1985;92:339-346.
sonication can be visualized directly as it is 4. Sanghvi NT, Fry FJ, Bihrle R. Noninvasive
being delivered. Thus, not only can the actual surgery of prostate tissue by high-intensity
temperature change be measured directly, but focused ultrasound. IEEE Trans Ultrason Ferro-
its precise location can be mapped. This occurs electr Freq Contr 1996;43:1099-1110.
for every sonication delivered during the entire 5. Chapelon JW, Ribault M, Vernier F. Treatment of
treatment. In a recent study, we found that localised prostate cancer with transrectal high
intensity focused ultrasound. Eur J Ultrasound
100% of visualized sonications were mapped.
1999;9(1):31-38.
The procedure continues with multiple soni- 6. Vallancien G, Harouni M, Veillon B. Focused
cations being delivered under direct MR guid- extracorporeal pyrotherapy: feasibility study in
ance and with real-time MR thermometry. man. J EndouroI1992;6:173-180.
After a therapeutic dose has been delivered to 7. Sanghvi NT, Hynynen K, Lizzi FL. New devel-
the target volume, postprocedural imaging with opments in therapeutic ultrasound. IEEE Biomed
two planes of TI-weighted images is obtained. Eng 1996;15:83-92. High intensity focused ultra-
The patient is then taken from the room and sound. Exp Invest Endosc 1996;1994;4:383-
observed for 1 to 2 hours, and then discharged 395.
with an escort or family member to drive her 8. Hiller R, Weninger K, Putterman SI Effect of
home. Success of treatment in this phase of the noble gas doping in single-bubble sonolumines-
cence. Science 1994;266:248-250.
protocol is judged on safety and feasibility. To
9. ter Haar G. Ultrasound focal beam surgery.
date, early experience indicates that the proce- Ultrasound Med Bioi 1995;21(9):1089-1100.
dure meets both criteria. 10. Crum LA, Hynynen K. Sound therapy. Physics
Ultimately the success of MRg FUS of World 1996;9:28-33.
uterine fibroids will be judged based on 11. Lehmann JF, DeLateur BJ, Warren CI Heating
improvement of the patient's symptoms when produced by ultrasound in bone and soft tissue.
this treatment modality is applied as a single Arch Phys Med Rehabil 1967;48:397-401.
21. Focused Ultrasound for Tumor Ablation 283

12. Hutchinson EB, Hynynen K. Intracavitary 27. Fry WJ, Barnard JW, Fry FJ. Ultrasonically pro-
phased arrays for non-invasive prostate surgery. duced localized selective lesions in the central
IEEE Trans Ultrason Ferroelectr Freq Contr nervous system. Am J Phys Med 1955;34(3):413-
1996;43:1032-1042. 423.
13. Hunt Jw. Principles of ultrasound used for 28. Lele PP. A simple method for production of
generating localized hyperthermia. In: Field SB, trackless focal lesions with focused ultrasound:
Franconi C, eds. Physics and Technology of physical factors. J PhysioI1962;160:494-512.
Hyperthermia. Boston: Martinus Nijhoff, 1987: 29. Heimburger RF. Ultrasound augmentation of
354-389. central nervous system tumor therapy. Indiana
14. Fjield T, Hynynen K. The combined concentric- Med 1985;78(6):469-476.
ring and sector-vortex phased array for MRI 30. Gelet A, Chapelon JY, Bouvier R. Urology
guided ultrasound surgery. IEEE Trans Ultrason Department, Edouard Herriot Hospital and
Ferroelectr Freq Contr 1997;44:1157-1167. INSERM Unite 28, Lyon, France.
15. Dorr LN, Hynynen K. The effect of tissue 31. Hill CR, ter Haar G. Review article: high-
heterogeneities and large blood vessels on the intensity focused ultrasound-potential for
thermal exposure induced by short high power cancer treatment. Br J Radiol 1996;68:1296-
ultrasound pulses. Int J Hyperthermia 1992;8(1): 1303.
45-59. 32. Gianfelice DC. MR guided focused ultrasound
16. Yang R, Sanghvi NT, Rescorla FJ. Liver cancer ablation of primary breast neoplasms: works in
ablation with extra-corporeal high-intensity progress. Radiology 1999;213:106-107.
focused ultrasound. Eur UroI1993;23:15-22. 33. Jolesz FA, Jakab PD. Acoustic pressure wave
17. Kolios MC, Sherar MD, Hunt Jw. Blood flow generation within an MR imaging system: poten-
cooling and ultrasonic lesion formation. Med tial medical applications. J Magn Reson Imaging
Phys 1996;23:1287-1298. 1991;1(5):609-613.
18. Moritz AR, Henriques FC Jr. Studies of thermal 34. Cline HE, Schenck JF, Hynynen K. MR-guided
injury. II. The relative importance of time focused ultrasound surgery. J Comput Assist
and surface temperature in the causation of Tomogr 1992;16(6):956-965.
cutaneous burns. Am J Pathol 1947;23:695- 35. Hynynen K, Darkazanli A, Unger E, Schenck JF.
720. MRI-guided noninvasive ultrasound surgery.
19. Crile G. The effect of heat and radiation on Med Phys 1993;20(1):107-115.
cancers implanted on the feet of mice. Cancer 36. Cline HE, Schenck JF, Watkins RD. Magnetic
Res 1963;23:372-380. resonance-guided thermal surgery. Magn Reson
20. Sapareto SA, Dewey WC. Thermal dose deter- Med 1993;30(1):98-106.
mination in cancer therapy. Int J Radiat Oncol 37. Jolesz FA, Hynynen K. Magnetic resonance
Bioi Phys 1984;10(6):787-800. image-guided focused ultrasound surgery.
21. Landry J, Marceau N. Rate-limiting events in Cancer J 2002;8(Suppll):SI00-112.
hyperthermic cell killing. Radiol Res 1978;75: 38. Hynynen K, Vykhodtseva NI, Chung AH.
573-585. Thermal effects of focused ultrasound on the
22. Delon-Martin C. Vogt C, Chigner E. Venous brain: determination with MR imaging. Radiol-
thrombosis generation by means of high- ogy 1997;204(1):247-253.
intensity focused ultrasound. Ultrasound Med 39. Chung AH, Jolesz FA, Hynynen K. Thermal
Bioi 1995;21(1):113-119. dosimetry of a focused ultrasound beam in vivo
23. Hynynen K, Colucci V, Chung A. Noninvasive by magnetic resonance imaging. Med Phys 1999;
artery occlusion using MRI-guided focused 26(9):2017-2026.
ultrasound. Ultrasound Med Bioi 1996;22(8): 40. McDannold NJ, King RL, Jolesz FA. Usefulness
1071-1077. of MR imaging-derived thermometry and
24. Vaezy S, Marti R, Mourad P. Hemostasis using dosimetry in determining the threshold for tissue
high intensity focused ultrasound. Eur J Ultra- damage induced by thermal surgery in rabbits.
sound 1999;9(1):79-87. Radiology 2000;216(2):517-523.
25. Moonen C, Madio P, de Zwart J. MRI-guided 41. McDannold NJ, Hynynen K, Wolf D. MRI eval-
focused ultrasound as a potential tool for control uation of thermal ablation of tumors with
of gene therapy. Eur RadioI1997;7:1165. focused ultrasound. J Magn Reson Imaging
26. Lynn Ja. A new method for the generation and 1998;8(1):91-100.
use of focused ultrasound in experimental 42. Hazle JD, Stafford RJ, Price RE. Magnetic
biology. J Gen PhysioI1942;179-193. resonance imaging-guided focused ultrasound
284 C. Tempany et al

thermal therapy in experimental animal models: 57. Cote I, Jacobs P, Cumming D. Work loss associ-
correlation of ablation volumes with pathology ated with increased menstrual loss in the United
in rabbit muscle and VX2 tumors. J Magn Reson States. Obstet GynecoI2002;100(4):683-687.
Imaging 2002;15(2):185-194. 58. Marshall LM, Spiegelman D, Barbieri RL. Vari-
43. Hynynen K, Pomeroy 0, Smith DN. MR ation in the incidence of uterine leiomyoma
imaging-guided focused ultrasound surgery of among premenopausal women by age and race.
fibroadenomas in the breast: a feasibility study. Obstet GynecoI1997;90(6):967-973.
Radiology 2001;219(1):176-185. 59. Baird D, Dunson D, Hill M. Cumulative inci-
44. Huber PE, Jenne JW, Rastert R. A new nonin- dence of uterine leiomyoma in black and white
vasive approach in breast cancer therapy using women: ultrasound evidence. Gen Obstet
magnetic resonance imaging-guided focused GynecoI2003;188(1):100-107.
ultrasound surgery. Cancer Res 2001;61(23): 60. Kjerulff KH, Langenberg P, Seidman JD.
8441-8447. Uterine leiomyomas. Racial differences in
45. Tempany CM, Stewart EA, McDannold N. MR severity, symptoms and age at diagnosis. J
imaging-guided focused ultrasound surgery of Reprod Med 1996; 41(7):483-490.
uterine leiomyomas: a feasibility study. Radiol- 61. Myers ER, Barber MD, Gustilo-Ashby T. Man-
ogy 2003;226:897-905. agement of uterine leiomyomata: what do we
46. Chen L,Bouley D, Yuh E. Study of focused ultra- really know? Obstet GynecoI2002;100(1):8-17.
sound tissue damage using MRI and histology. 62. Bonney V. The technique and results of myomec-
J Magn Reson Imaging 1999;10(2):146-153. tomy. Lancet 1931;1:171-177.
47. Hynynen K, Darkazanli A, Damianou CA. The 63. Ecker JL, Foster FS, Friedman Al Abdominal
usefulness of a contrast agent and gradient- hysterectomy or abdominal myomectomy for
recalled acquisition in a steady-state imaging symptomatic leiomyoma: a comparison of pre-
sequence for magnetic resonance imaging- operative demography and postoperative mor-
guided noninvasive ultrasound surgery. Invest bidity. J Gynecol Surg 1995;11(1):11-18.
Radiol 1994;29(10):897-903. 64. Iverson RE Jr, Chelmow D, Strohbehn K. Rela-
48. Graham SJ, Stanisz GJ, Kecojevic A, Bronskill tive morbidity of abdominal hysterectomy and
MJ, Henkelman RM. Analysis of changes in MR myomectomy for management of uterine
properties of tissues after heat treatment. Magn leiomyomas. Obstet Gynecol 1996;88(3):415-
Reson Med 1999;42(6):1061-1071. 419.
49. Rowland 11, Rivens I, Chen L. MRI study of 65. Stewart EA, Faur AV, Wise LA. Predictors of
hepatic tumours following high intensity focused subsequent surgery for uterine leiomyomata
ultrasound surgery. Br J RadioI1997;70:144-153. after abdominal myomectomy. Obstet Gynecol
50. Stewart EA. Uterine fibroids. Lancet 2001;357: 2002;99(3):426-432.
293-298. 66. Spies JB, Roth AR, Jha RC. Leiomyomata
51. Law P, Gedroyc WM, Regan L. Magnetic treated with uterine artery embolization: factors
resonance-guided percutaneous laser ablation associated with successful symptom and imaging
of uterine fibroids. J Magn Reson Imaging 2000; outcome. Radiology 2002;222(1):45-52.
12(4):565-570. 67. Pron G, Bennett J, Common A. The Ontario
52. Law P, Gedroyc WM, Regan L. Magnetic reso- uterine fibroid embolization trial part 2. Uterine
nance guided persutaneous laser ablation of fibroid reduction and symptom relief after
uterine fibroids. Lancet 1999;354:2049-2050. uterine artery embolization for fibroids. Fertil
53. Sewell PE, Arriola RM, Robinette L. Real-time SteriI2003;79(1):120-127.
IMR-imaging-guided cryoablation of uterine 68. Goodwin SC, Walker WI Uterine artery embo-
fibroids. J Vasc Interv RadioI2001;12:891-893. lization for the treatment of uterine fibroids.
54. Carlson KJ, Nichols DH, Schiff I. Indications Curr Opin Obstet GynecoI1998;10(4):315-320.
for hysterectomy. N Engl J Med 1993;328(12): 69. Vashisht A. Studd J, Carey A. Fatal septicaemia
856-860. after fibroid embolisation. Lancet 1999;354
55. ACOG practice bulletin. Surgical alternatives to (9175):307-308.
hysterectomy in the management of leiomy- 70. Godfrey CD, Zbella EA. Uterine necrosis after
omas. Int J Gynaecol Obstet 2001;73(3):285- uterine artery embolization for leiomyoma.
293. Obstet GynecoI2001;98(5 Pt 2):950-952.
56. Spies JB, Ascher SA, Roth AR. Uterine artery 71. Spies JB, Spector A, Roth AR. Complications
embolization for leiomyomata. Obstet Gynecol after uterine artery embolization for leiomy-
2001;98(1):29-34. omas. Obstet GynecoI2002;100(5 Pt 1):873-880.
22
New Technologies in Tumor Ablation
Bradford 1. Wood, Ziv Neeman, and Anthony Kam

The technology and engineering of tumor deposition. Both temperature and impedance
ablation are evolving more rapidly than the reflect this process. The logarithmic increase in
clinical validation. Technical descriptions risk impedance that accompanies early overcooking
being obsolete by publication time. General and charring is accompanied by an automated
principles and developing technical paradigms drop in current to allow tissue to cool (pulsing).
provide a simplified framework to practice This avoids the "char-broiled burger effect," in
and study radiofrequency ablation (RFA). This which the tissue in immediate contact with the
chapter reviews the general limitations of tissue electrode is overcooked, but the meat further
ablation methods, the optimization of ablation from the heat source is raw. The goal should be
in the radiofrequency range, and several emerg- to cook as evenly as possible to allow uniform
ing technologies and paradigms feasible in the thermal conduction throughout the soft tissues.
laboratory and possibly translatable to clinical The limitations of various ablation methods
practice. can be divided into three categories: ablation
volume, ablation shape, and lesion targeting.
The first of these has received 95% of attention
to date. The theme of the first decade of RFA
General Principles for tumors, the 1990s, was "bigger is better."
Most of the developments had the goal of
Most efforts in the early development of larger tissue destruction in shorter times. This
ablation systems focused on altering the needle was largely successful, with 7-cm shapes being
configuration, tissue properties, or treatment attained in tens of minutes. However, the pre-
algorithms to optimize volume of tumor dictability and reproducibility of ablation size
kill. Early technical developments reflected and shape have received little attention. In
efforts to improve relatively similar RFA reality, the shapes of the thermal lesions are not
systems. These modifications include deploy- spherical or completely predictable. This com-
able probes, hypertonic saline infusion, multi- plicates geometric overlapping, increases risk
probe synergy, chilled electrodes, and current for collateral damage, and worsens outcomes.
pulsing. Although vendors describe various The different systems create different shapes:
benefits of their respective systems in market- oval, teardrop, or disk-like (Fig. 22.1). Custom
ing, temperature and impedance control are shaping of the thermal lesion with microwave,
highly interrelated and similar. As temperature bipolar RFA, or multiprobe systems may be
approaches boiling (100°C), tissue charring, the next large technical development in tissue
vaporization, and a coalescence of organic ablation. Custom thermal lesion shaping might
microbubbles at the electrode surface all inter- lead to improved outcomes, with less collateral
act to insulate the electrode and limit energy damage to normal structures.

285
286 B.J. Wood et al

FIGURE 22.1. Thermal lesion size and shape in rela- varies significantly among vendors [Radionics
tion to needle geometry is an area that needs further (Radionics Inc, Burlington, MA), RITA (RITA
clarification. Precise needle placement in relation to Medical Inc., Mountain View, CA), RadioTherapeu-
resulting thermal lesion shape and tumor geometry tics (Boston Scientific, Natick, MA)].

Other Ablation Technologies source of heat to propagate peripherally within


a thermal lesion. This mechanism may over-
Tumor size is the most important factor in come the "heat-sink effect", as tissue perfusion
determining complete response rates for the has less time to cool down adjacent tissue.
various local or regional cancer therapies. In high-intensity focused ultrasound (HIFU),
This has led to the principle that for ablation each application of an ultrasound pulse pro-
volume, bigger is better. In alcohol ablation, the duces a unit coagulation volume similar in
ablation volume is determined by the amount size to a grain of rice. The ablation volume is
of alcohol that is injected. Although there is dependent on the ability to overlap these unit
no safe cut-off volume of alcohol that can be volumes, the length of the procedure, and res-
injected, in practice, this volume is limited by piratory and patient motion. Although there
systemic and local toxicity. In radiofrequency are many choices for electromagnetic energy
and laser ablation, direct heating takes place deposition in tissue, future ideal tissue ablation
within a few millimeters of the electrode or technology will have to surpass RFA in ease
fiber; heating at a distance from the electrode of use, convenience, cost, safety, accuracy,
or fiber occurs via conduction. The probe itself predictability, reproducibility, practicality, and
does not heat up primarily, but does so secon- efficacy.
darily from heat conducted from adjacent
tissue. The ablation volume is primarily limited
by cooling from tissue perfusion. Microwave
Heat Transfer
ablation shares this limitation except that Much early work in tissue ablation focused on
direct heating extends deeper into tissue. Thus, optimization of heat transfer: increasing energy
microwave ablation has the advantage of being deposition, improving tissue heat conduction,
less dependent on thermal conduction, since a and minimizing heat loss. This is within the
large volume of tissue receives energy rapidly, framework of the oversimplified view of
and unlike RFA, does not depend on a narrow Penne's classic bioheat equation: coagulation
22. New Technologies in Tumor Ablation 287

necrosis is equal to energy deposited minus is complicated by the relative effects of small
heat lost. The bioheat equation is more useful and large vessels, flow rates, and countercurrent
if it is quantitatively solved to predict ablation heat exchange of warmed vessels.
volumes and shapes for various clinical situa-
tions. Deciding where to place a probe or ex- Tissue Dielectrics
actly when to use an occlusion balloon in a
The dielectric correlates of specific tumor and
specific clinical situation may be helped by
organ histologies also are poorly characterized.
these developing mathematical models. For
Few data are published that differentiate organ
instance, if a 3-mm vessel lies within 5 mm of a
and tumor effects. Identical RFA algorithms
tumor, modeling may be helpful to decide if
and parameters may result in surprisingly dis-
balloon occlusion of the vessel is necessary to
parate ablation volumes in different locations,
ablate the tumor.
independent of perfusion (Fig. 22.2). Adreno-
Tumor modeling requires knowledge of the
cortical carcinoma, lymphoma, and chordoma
input parameters of the bioheat equation that
are three cell types we have noticed that
include density, specific heat, thermal conduc-
repeatedly heat quicker than usual, for
tivity, electrical conductivity, and perfusion of
example. The easier RFA of hepatocellular
tissue. These values are not available for most
carcinoma versus same-size colorectal liver
tumors. Instead, the values from the organ in
metastases also can be explained partially by
which the tumor resides are extrapolated. Also,
differing dielectrics. Identifying the thermal
the values used in the various models are at body
and electrical conductivities of certain organs
temperature. The temperature coefficient of
or tumors within clinical ranges of temperature
tissue electrical conductivity above 40°C is sub-
and frequency could lead to tissue-specific and
stantially different from the coefficient below
organ-specific algorithms that could improve
40°C at frequencies used in RFA. Current mod-
outcomes. For example, a different algorithm
eling is based on false assumptions using con-
and set of rules are used for kidney tumor abla-
stants that are true at body temperature, but are
tion. The resulting volumes are predictably
not true for tissue in the 50° to 100°C range.
smaller than for liver, although this is largely
Finally, when blood vessels are modeled, it has
perfusion-dependent. Ablation generators may
been assumed that the temperature of blood is
be made with removable tissue and organ-
body temperature. This may not be valid for
specific modules that reprogram treatment
small blood vessels. The utility of validated
algorithms for specific tissue and organ types.
models will be immense as more complex and
larger tumors are considered for RFA.
Analysis of reflected versus absorbed power
Tumor Environment
could shed light on the variability of RFA. The effects on RFA of high interstitial pres-
Finite element modeling has been applied to sures within a tumor center, or an acidic or
various electrodes in an attempt to better hypoxic environment are unknown. Radiofre-
understand and predict tissue effects (1). Per- quency ablation likely is less affected by the
sonalized treatment planning and automated rugged environment in the typically poorly
probe placements are being integrated into perfused tumor center. As such, RFA is an
RFA treatments. There is a poor understanding ideal candidate for combination therapies with
of the nonlinear function of thermal and elec- blood-borne adjuvants like intravenous (IV)
trical conductivities in the clinical range (50° to chemotherapy, IV liposomal vectors, chemoem-
lOO°C near 500kHz). Heat transfer is by con- bolization, or directly injected adjuvants like
duction in solids, and by convection in fluids or polymerized drug depots or chemotherapy gels.
flowing blood. Radiofrequency ablation works Intravenous chemotherapy works on rapidly
by alternating current; thus there is a complex dividing cells at the tumor periphery where
interaction between electrical and thermal there is good blood supply, and RFA works
forces. The mathematical description of the per- on the highly pressurized and devascularized
fusion effect (loss of heat due to convection) center of a tumor. Combination therapies like
288 B.J. Wood et al

FIGURE 22.2. Radiofrequency ablation (RFA) burn with similar parameters and same histology. We
variability. The smaller renal cell carcinoma burn on know little about the tissue effects of RFA, and it is
the left (arrow) was made by 24 minutes of treat- not as predictable as marketed.
ment, and the larger burn on the right by 3 minutes

RFA plus chemoembolization or balloon occlu- probes, coaxial probes, and tissue-specific
sion take advantage of such complementary probes (bone). Further refinements could
effects; however, they likely add risk. The exact include biodegradable tapered tips to prevent
timing, sequence, and indications for these capsule coring, and spring deployment to
combination therapies have not been defined ensure complete deployment and avoid "kick-
or optimized. back" of the outer cannula that can result in
misshaped deployments. Special probes could
be developed to optimize the heat profiles for
specific combination therapies. Specifically, new
Technology for Optimization probes could leverage the benefits of heat
of Treatment Planning activated drug delivery, drug depot polymers,
and Monitoring or enhanced permeability (patents pending).

Imaging
Imaging also is an imperfect surrogate for tumor
Radiofrequency Ablation
and Thermometry
margins with indistinct, irregular, or infiltrating
edges. Better imaging with ultrasound contrast Percutaneous image-guided RFA risks thermal
agents, three-dimensional ultrasound, multipla- damage to nearby normal tissues. For tumors
nar reconstruction, image processing, elastic near sensitive anatomy, there is a trade-off
fusion, dynamic MR, computed tomography between aggressive treatment that risks com-
(CT), fluoroscopy, noninvasive thermography, plications and undertreatment (which risks
and MR thermometry also have been the residual tumor). Thermometry may help the
subject of intense research. operator negotiate this trade-off. The exact
thermal lesion margin remains somewhat un-
predictable. Therefore, when temperature-
Equipment sensitive structures (such as bowel and nerves)
The rapid rate of technical developments has are nearby, direct (mounted on the RF needle)
fueled the revolution in tissue ablation. Recent or indirect (remote or independent) ther-
developments include a flexible probe that mometry could further increase the safety of
can fit into a CT gantry, MR-compatible RFA.
22. New Technologies in Tumor Ablation 289

To date, thermometry has been described thermometry may provide noninvasive, real-
with a multitude of low-temperature hyper- time monitoring of coagulation during HIFU,
thermia systems, as well as with prostate abla- microwave, hot saline, laser, and RFA proce-
tion. Different tissues have variable thermal dures. This is possible because of the tempera-
thresholds. This underscores the importance of ture dependence of spin-lattice relaxation time,
real-time intraprocedural thermometry near proton resonance frequency, and diffusion.
adjacent, susceptible organs. For example, the The disadvantages are inaccurate temperature
temperature threshold for bowel injury when monitoring of moving targets due to respira-
ablating the prostate is 45°C; myocardial injury tion, scarce availability, switching back and
occurs at 50°C; and nerve pain fiber ablation forth from MR RF pulses to RFA, and the cost
occurs at 45°C. We routinely use remote ther- of MRI-compatible RFA systems. Also, the
mistors to protect adjacent organs during RFA, most robust sequences [proton resonance fre-
and we often modify the energy delivered or quency (PRF)] are also very susceptible to
the treatment duration to limit risk to collateral breathing motion artifact.
structures.

Robotics
Direct (Dependent) Thermistors Robotics is being applied rapidly to RFA to
Some available RFA systems incorporate ther- assist needle placement. For percutaneous
mistors to evaluate tissue temperatures at the image-guided procedures, the main advantage
tip of the active RFA probe during post-RFA of robotics is that it facilitates the use of a large
cooling such as Radionics (Radionics, Inc., amount of quantitative data from imaging;
Burlington, MA), or near the margin and in consequently, it allows a greater accuracy and
between active tips such as RITA (RITA precision in needle placement. A secondary
Medical Inc., Mountain View, CA). The down- advantage is that for CT or x-ray ftuoroscopic-
side of this technique is that the thermistors guided interventions, robotics can eliminate
cannot be deployed separately from the probe radiation exposure to the interventionalist (2).
itself. Although these thermistors are directed Machines are more accurate than humans
to the target tissue, they cannot provide pre- when it comes to planning and enacting multi-
cise temperature information about adjacent ple overlapping treatments. Robotics also may
nearby nontarget tissue. facilitate translation of needle-based technolo-
gies into the community setting.
Defining robotic language will help one
Indirect (Independent) Thermistors
understand how these machines will soon trans-
There are remote thermistor systems available. form how needles are placed. Bloom et al (3)
Radionics makes a 25-gauge accessory thermis- describe fundamental robotics terminology as
tor that acts as a stylet inside a 22-gauge SMK follows: A robotic arm consists of links that are
pole needle, with a digital TCA-2 monitor. The connected by joints. At the distal end, there
advantage is independent placement and posi- is an end-effector that performs the action of
tioning of the thermistors between the RFA the robot. The axes through which an arm
probe and collateral, at-risk structures. can translate or rotate define the degrees of
freedom for the arm. For instance, since there
are three degrees of translational freedom (x,y,
Magnetic Resonance Thermometry and z axes) and three degrees of rotational
Another way to monitor real-time temperature freedom around those axes (pitch, roll, and
changes during RFA is by MR thermometry. yaw), an end-effector that has six degrees of
This method gives three-dimensional volumet- freedom can take any position and any orien-
ric data, and thus more reliably indicates tem- tation. The terms active and passive have dif-
perature changes in the entire tissue of interest, ferent meanings depending on whether they
not just from one point. Magnetic resonance refer to a section of a robot or to the entire
290 B.J. Wood et al

robot. A passive section is one that has to be


moved manually; an active section is one that
is self-propelled. For the entire robot, active
and passive refer to the degree of autonomy.
Active robots perform a task according to
a program and without human assistance.
Semiactive robots require human interaction to
perform a task. Passive robots do not perform
any task, but supply information.
A group at Johns Hopkins constructed a
robotic arm for percutaneous interventions
(2,4). Called PAKY-RCM (percutaneous
access to the kidney-remote center of motion),
this robot consists of a passive arm with seven
degrees of freedom. Attached to this arm is an FIGURE 22.4. Active robot prototype demonstrating
seamless direct integration of CT and robot by
active remote center of motion that pivots a
mechanically registering the robot to the CT coordi-
needle about its tip. An active needle driver
nates using a gantry frame. (Copyright Jeff Yanof,
holds the needle. The system can be operated PhD, Philips Medical Systems, Cleveland, OR.)
as a semiactive robot (a surgeon manipulates
a joystick to drive the needle), an active robot
(completely computer-controlled with the intervention in 75% of patients. The remaining
robot indirectly registered to the CT image), or 25% were successful after fine adjustments
in combination. The Hopkins' group has shown with the joystick. Georgetown has developed a
that the number of attempts, time to access, and spinoff robot for fluoroscopic-guided needle
safety of robotic percutaneous access using interventions driven by a joystick (Fig. 22.3).
PAKY-RCM and standard manual percuta- We are helping to design an interventional
neous access of renal calyces under fluoroscopy suite with a robot prototype mechanically reg-
were comparable. For CT-guided interventions istered to CT using a mechanical frame and arm
involving the kidney, liver, neobladder, spine, (Philips Medical Systems, Cleveland, OH) (Fig.
lung, and muscle, this group has achieved 22.4). During the first attempt with the proto-
success at hitting the target without human type, a I-mm BB was hit with a 22-gauge needle
on a complex oblique path 15 cm deep in the
dome of a phantom liver without CT fluo-
roscopy (5). Free-hand cannot be as accurate
and precise.

Navigation, Needle Tracking,


Image Fusion
Seamless integration with the imaging data
set is a necessity for successful robotic-assisted
RFA. Safety issues, respiratory motion, and
organ shift during procedures remain the
largest obstacles to robotic deployment for
organs other than the brain, spine, prostate, and
bone. The same obstacles limit utility of navi-
FIGURE 22.3. Table-mounted robot integrated with gation and augmented reality systems to guide
biplane fluoroscopy. Needle is driven using joystick RFA. Magnetic tracking may be the most
controlled by physician. (Courtesy of Kevin Cleary, promising navigational tool for RFA (Figs. 22.5
PhD, Georgetown University.) to 22.7). A weak magnetic field is generated
FiGURE 22.5. Conventional magnetic tracking device FIGURE 22.6. Conventional magnetic tracking
with clips on the ultrasound and the needle shaft. device showing out of plane needle navigation with
ultrasound.

FIGURE 22.7. (A) Magnetic


field generator for intersti-
tial magnetic tracking with
small sensors inside needle
shaft. (B) Graphical user
interface for magnetic
tracking and sphere-
packing for planning and
navigating RF ablation.
(Courtesy of Kevin Cleary,
PhD, Georgetown
University.) B
292 B.J. Wood et al

A B
FIGURE 22.8. (A) Volumetric display of fused pre- lesion 1 month post-RFA to define targets of resid-
and post-RFA images shows residual tumor and its ual tumor for next RFA. (Courtesy of Frederik
spatial relationship to thermal lesion. (B) Rigid Geisel, Hendrik von Tengg-KobIigk, Matt McAuliffe,
image fusion shows positron emission tomography and Medical Image Processing and Visualization
(PET)-positive tissue at the edge of the thermal Program, NIH.)

and small sensors may be placed on the ultra- re-treated before becoming geometrically
sound transducer, the RFA needle, as well as unfavorable. Using a computer with Java-based
the patient. The exact x,y,z position in space image processing software, RFA can be per-
and the projected pathway" are displayed to formed with online real-time semiautomated
assist needle placement. Major shortcomings image fusion to define targets and identify
thus far have been related to the position of the residual untreated neoplastic tissue (Fig. 22.8).
needle-based sensor, which is clipped to the Combined PET-CT scanners are popularizing
needle shaft/hub junction in one common com- the same process for diagnostic purposes.
mercialized system (Figs. 22.5 and 22.6). This is
prone to misregistration with bending of the
needle, organ shift, or respiratory variation. Emerging Paradigms:
Efforts are under way to optimize the use of Radiofrequency Ablation as
interstitial sensors within stylets or RFA probes
or internalized fiducials to optimize position
Adjuvant Therapy?
information (Fig. 22.7) (patent pending).
The difficulties of organ shift and respiratory
Gene Therapy
motion also complicate multiparametric image Gene therapy has a long way to go to be clini-
display or image fusion as guides for RFA. cally relevant and mainstream. However, when
Image fusion may take advantage of off-line it arrives, it may be combined with heat, either
modalities during aCT-guided RFA. Pretreat- as an adjuvant or as heat-activated gene deliv-
ment CT, MR, or positron emission tomogra- ery. This could be accomplished with a heat-
phy (PET) may be fused with the procedural activated particle or with a heat-upregulated
imaging or the follow-up imaging to facilitate gene promoter. ThI:1 heat shock protein pro-
early detection of residual tumor so it may be moter has been u~ed in animal studies to
22. New Technologies in Tumor Ablation 293

deliver genes locally to heated tissue. Gene Radiation and Radiofrequency


delivery is verified by reporter gene (green Ablation
fluorescent protein) imaging (6). Cleavable
linkers also may be heat activated, releasing the Hyperthermia is a useful adjunct to radiother-
active moiety upon exposure to heat (7). DNA apy for the local control of advanced malig-
may be engineered in the laboratory to manip- nancies. Low-temperature hyperthermia may
ulate tumor biology or the cellular response be the most effective cellular radiosensitizer
to therapies like heat and chemotherapy. known. Heat acts via several pathways. Heat
Genes under investigation for cancer include alone independently kills radioresistant cells
therapeutic genes, oncogenes, suppressor genes, (s-phase and hypoxic cells). Heat also poten-
suicide genes, antisense genes, reporter genes, tiates the adverse effects of radiation on the
plasmid DNA, co-stimulatory genes, and genes cell's ability to repair radiation-induced DNA
coding for drug resistance, cytokines, antigens, damage (10). These effects occur throughout
drug efflux pump, and immunomodulation, to the entire cell cycle, but most prominently at
name but a few. The main obstacles to in vivo the s phase.
gene therapy have been specific and efficient Although the exact timing and sequence
delivery, transfer, and expression of the vector. remain to be optimized for human RFA plus
Toxicity and inadequate targeting are related radiation, there is much to be learned from
impediments. prior experience in lower temperature hyper-
Other forms of electromagnetic energy out- thermia plus radiation. In rodent studies, the
side the SOO-kHz range may be more effective highest level of radiosensitization was achieved
at promoting gene transfection or altering local when hyperthermia was applied simultaneously
permeability. Ultrasound may augment gene with radiation (with a radiosensitization half-
transfection efficiency in vivo (8). The effects life of 20 to 30 minutes). In humans, there is
of sublethal RFA upon gene transfection are likely a 4-hour window (radiosensitization half-
undefined. By the time gene therapy has life of 100 to 120 minutes).
moved beyond the mouse, in an effective clini- The synergistic effects of radiation plus
cal trial, tissue ablation systems will likely be hyperthermia have been supported by several
completely different from the way they are cur- recent reports, with various successful random-
rently. ized clinical trials, including recurrent advanced
breast cancer, recurrent or metastatic mela-
noma, head and neck cancer, non-smaIl-cell
Immunotherapy lung cancer, glioblastoma multiforme, and intra-
Thermal ablation with microwaves has been pelvic malignancies (bladder, uterine cervix,
shown to stimulate a tumor-specific immune and rectum). Ex-vivo studies suggest that most
response remote from the heated tissue (9). cell lines are sensitive to this synergistic com-
Radiofrequency ablation may act in a similar bination therapy.
fashion, and may improve the function of While the additive effects of hyperthermia
dendritic cells or the presentation of tumor and radiation have been well studied at low
antigens to the immune system, as it recruits temperatures (40-44°C), there is sparse infor-
macrophages to the periphery of the thermal mation regarding radiation and the RFA tem-
lesion in the sublethal zone (7). This immune perature range (60-100°C). Heat may im-
response may be redirected to fight circulat- prove the complex relationship among local
ing tumor in remote locations. High-intensity perfusion, interstitial pressure, and oxygen
focused ultrasound may hypothetically be the tension in the center of an irradiated tumor.
best antigen-presenting technology, given the Radiation effects on perfusion could reduce
violence inherent in cavitation, whereby tumor heat sink convection during RFA, which could
antigens may be presented suddenly and augment ablation volumes. The synergism
forcefully. Preclincial work needs to be done to between RFA and radiotherapy needs further
answer these questions. clarification.
294 B.1. Wood et al

Drug Delivery
Radiofrequency ablation is one of several
ways to deliver heat, with the advantage of
selectivity by allowing heat to be delivered
more specifically to the local tumor. Also, RFA
may prove to be a tool broadly used for drug
delivery or therapy augmentation. In this para-
digm, RFA debulks the tumor and potentiates
the local or regional effects of the combination
therapy. Antiangiogenic targeted therapies may
inhibit and repress a tumor without complete
cell death. In this fashion, some cancers in the
future may be kept from growing or metasta-
sizing without complete eradication. Given that
cancer may be considered a chronic disease in
the future, RFA debulking also may evolve FIGURE 22.10. A combination of therapies, delivery
into a more accepted and standard rational systems, and strategies may be more effective than
practice. However, this may require translation single therapy. RFA plus chemoembolization, in-
of antiangiogenic therapies from the bench to jectables, gene therapy, molecular targeted drugs,
the bedside. Debulking theoretically may allow liposomes, or immunotherapy should open doors to
other therapies to work better against smaller more broad clinical applications for thermal ablation
in the future. (Courtesy of Jay Patti, MD.)
volumes of disease, although this is controver-
sial in the oncology community, and largely
depends on the cell type and clinical scenario.
Aggressive debulking is more commonly being
performed when the natural history of a neo-
plasm has proven itself to be less aggressive
over time.
Selective drug delivery in cancer has met
with limited success thus far due to a combina-
tion of problems. These include getting to the
cell, getting inside the cell, staying there,
avoiding natural defense systems, and minimiz-
ing systemic toxicity and effects on normal
tissue. Radiofrequency ablation may be used to
augment drug delivery via heat-activated parti-
cles or by simple increased permeability from
sublethal heating (Figs. 22.9 and 22.10).

Drug Depot Effect


Tumor cell microenvironment may be made
favorable for drug uptake or effect. Cryother-
apy increases bleomycin uptake in tumors (11).
Cryotherapy also increases membrane perme-
FIGURE 22.9. Selective hepatic artery angiography ability. Any ablation technology potentially
shows hypervascular rim (white arrow) around increases drug dwell time due to devasculariza-
thermal lesion (black arrow) immediately following tion and vascular stasis. Radiofrequency abla-
RFA. tion may further facilitate the drug depot effect,
22. New Technologies in Tumor Ablation 295

where the devascularized zone of coagulation


necrosis holds on to the drug for much longer
than in perfused tissue. In this model, the drug
slowly seeps out of the coagulation necrosis
sponge over time, resulting in lasting drug dwell
time. Drug washout by perfusion is overshad-
owed by drug diffusion into surrounding tissues.
This concept has been postulated by Haaga's
group (12) with implanted doxorubicin-eluting
polymer implants in rabbit liver following RFA.
Drug-eluting millirod polymers combined with
RFA could represent a winning combination
that warrants further resources and study.

Electroporation
Drug, gene, particle, or protein delivery to the
cell may be facilitated with electroporation
or sonoporation. Electroporation therapy is
the more mature of the two. In this technique,
pulsed electric fields are used to temporarily
increase the permeability of cell membranes
by creating transient pores (13). Extracellular
macromolecular therapeutic agents like drugs,
proteins, or genes that normally have difficulty
crossing cell membranes can enter cells through
these pores. Needle electrodes placed directly FIGURE 22.11. Intratumoral electroporation system
into the target tissue are used to apply the for enhanced drug and gene delivery. (Photo printed
pulsed electric fields. As a method for drug or with permission from Genetronics, Inc.)
gene delivery for cells in vitro, electroporation
is well established. Electroporation also pro-
longs retention of plasmid-mediated gene Genetronics Inc. (San Diego, CA) is currently
transfer in liver vasculature (14). sponsoring a phase III clinical trial of elec-
Electroporation-assisted gene therapy trochemotherapy with intratumoral bleomycin
(electrogene therapy) currently remains in the for head and neck cancers (Fig. 22.11). Sersa
preclinical stage; however, electroporation- et al (16) reported a phase II trial involving elec-
assisted chemotherapy (electrochemotherapy) trochemotherapy with intratumoral cisplatin
has reached the stage of clinical trials. One for malignant melanoma; 133 tumor nodules in
advantage of electrochemotherapy over stan- 10 patients were divided as follows: 82 nodules
dard chemotherapy is that side effects are min- were treated with electrochemotherapy, 27
imized because a much lower dose of the drug nodules were treated with intratumoral cis-
is used. One group of such trials involves elec- platin, two nodules were treated with electric
trochemotherapy with bleomycin. In 50 pa- pulses alone, and 22 nodules were not treated.
tients with 291 cutaneous or subcutaneous After 4 weeks, objective (complete and partial)
malignant tumors treated with e1ectroporation response was achieved in 78 % of nodules in the
and bleomycin given interstitially or intra- electrochemotherapy group, whereas objective
venously, Mir et al (15) reported a complete response was achieved in 38% of nodules in the
response in 56.4 % of tumors and a partial intratumoral cisplatin group.
response in 28.9% of tumors, with minimal side One barrier in the development and opti-
effects. mization of electroporation therapy in vivo is
296 B.J. Wood et al

that currently there is no method to monitor solid vibrator. The microstreaming has a sharp
the electroporation process in real time. Param- velocity gradient that causes a shear stress on
eters for electric field pulses (amplitude, the solid surface or, in the present context, the
duration, duty cycle, and number) are typically cell membrane. The shear stress stretches the
based on preclinical work. For instance, micro- cell membrane outward and partially damages
second pulses with high electric fields are used it.
in electrochemotherapy, and millisecond pulses Understanding the factors that affect cavita-
with lower electric fields are used in elec- tion is an area of active research. A few obser-
trogene therapy (17). Necrosis and tumor vations can be made. Cavitation requires the
shrinkage or expression of a gene are used presence of cavitation nuclei in a media. In the
to assess the efficacy of the electroporation absence of such nuclei, inertial or spontaneous
process. Preclinical work has shown that for cavitation occurs with a threshold of acoustic
electrogene therapy with a fixed set of pulse pressure. In contrast to electroporation, there
parameters, transfection efficiency varies with has been less in vitro and preclinical work using
the tumor type (17). Consequently, preclinical sonoporation to enhance the delivery of drugs
pulse parameters are not necessarily optimal and genes (23,24). The frequencies used in
for the clinical arena. these studies are typically much lower than the
Recently, Davalos et al (18) proposed using frequencies used in diagnostic ultrasound, on
electrical impedance tomography (EIT), a non- the order of kilohertz.
invasive method of imaging the impedance
distribution inside the body, to monitor the
electroporation process. Multiple electrodes
Shock Waves and Ultrasound-
are placed on the body surface. Current is then
Released Particles
sourced at each electrode successively. Surface Shock waves may be used with similar goals.
potentials are measured with each current Shock waves are single pulses of pressure in
source. The impedance image is obtained by which the speed of propagation is greater than
solving Laplace's equation with the measured the speed of sound. Shock waves have more
boundary values. The hypothesis of Davalos et pressure and are more localized than typical
aI's proposal is that when pulsed electric fields ultrasound. Shock wave poration causes in-
are successful in creating pores in cell mem- creased chemotherapy uptake and increased
branes, there will be a measurable change in membrane permeability. Ultrasound-mediated
tissue impedance. particle delivery systems have been developed
in which the particle is burst locally by the
ultrasound, which then locally deploys the pay-
Sonoporation
load in a fashion similar to some ultrasound
Another method of increasing the permeability contrast agents. Because ultrasound can be tar-
of cell membranes transiently is ultrasound. geted and is noninvasive, ultrasonic technology
Unlike HIFU, sonoporation is not a thermal for drug delivery warrants further development
injury. Althqugh not well understood, sonopo- and is receiving much attention from the bio-
ration is believed to be mediated through technology industry currently.
cavitation. The evidence for this comes from
a series of in vitro experiments showing that
ultrasound contrast microbubbles enhance
Permeability
sonoporation (19,20). In an elegant experiment, Sublethal thermal ablation enhances perme-
Wu et al (21,22) have shown that microstream- ability and increases endothelial leakage, possi-
ing is responsible for sonoporation. Acoustic bly widening endothelial gap junctions. Studies
streaming is a steady flow in a liquid induced by of hyperthermia (40° to 50°C) show increased
an acoustic field. Microstreaming is a vortex- selective drug uptake and decreased micro-
like streaming that occurs next to an oscillating vascular flow (less washout). A temperature of
bubble near a solid surface or next to a small 41°C preferentially increases vascular perme-
22. New Technologies in Tumor Ablation 297

ability of tumor neovessels in a temperature-


dependent fashion (25).
Radiofrequency ablation kills tissue in the
center of a tumor where there are high inter-
stitial pressures, ischemia, hypoxia, and poor
perfusion. However, at the periphery of a
thermal lesion, there remains a rind of hyper-
vascular tissue (Fig. 22.9). This hypervascu-
larity with leaky vessels should potentiate
chemotherapy deposition to the cells that are
most likely to be incompletely heated. This
could allow for chemotherapy to reach residual
viable actively dividing cells, where it may be
most effective. Also, more cells in the periphery
of a tumor generally will be undergoing cell
division, and many chemotherapies work by
this process.
Cryotherapy increases intracellular chemo-
therapy deposition in cells undergoing sub-
lethal freezing, likely via increased membrane
permeability and trapping of drug from vascu-
lar stasis. Radiofrequency ablation should have
a similar drug depot effect, because there is
increased membrane fluidity and increased
endothelial leakage (26). Focused ultrasound
also may prove useful for increasing drug deliv-
ery via a thermal effect as well as mechanical
shaking of the tissue and vasculature with a
high-amplitude subsecond pulsed wave (27).

Thermally sensitive
Liposomes/Nanoparticles deployment, surface
associated targeting
Drug-laden liposomes take advantage of this information
altered permeability to improve drug de- (Monoclonal antibody
position. First-generation liposomes have or ligand)
phospholipids that encapsulate drugs. Second-
generation liposomes incorporate polyethylene
glycol (PEG) to stabilize the particle and result FIGURE 22.12. Evolution of liposome technology for
in prolonged circulation and less phagocytosis drug delivery. Polyethylene glycol (PEG) coating
or reticuloendothelial system uptake (Fig. results in prolonged circulation and avoidance of
22.12). Early laboratory and clinical work phagocytosis. Liposomes are a rational choice for
suggest that such PEG-ylated liposomes en- RFA heat-activated therapeutics.
capsulating a payload of doxorubicin (Doxil/
stealth liposome) may be potent adjuvants for The dosing has not yet been optimized, how-
RFA (28). Polymerization and polyvalency of ever. Third-generation liposomes incorporate
liposomes can improve delivery of standard targeting ligands or monoclonal antibodies
chemotherapy liposomes, with a high payload, for specific delivery and local/regional effect
low toxicity, and prolonged circulation. High (Fig. 22.12). Early laboratory studies suggest
heating of liposomes also may release cytotoxic that a low-temperature-sensitive liposome en-
free radicals that potentiate cytoreduction. gineered at Duke University (Durham, NC)
298 B.1. Wood et al

may be advantageous for combining with RFA.


This liposome deploys its cargo much faster Targeting antibody or ligand

than other liposomes (on the order of seconds)


and then seals up after passing the heated tissue
from RFA. It also is much more thermally sen- Encapsulated payload

sitive than other conventional doxorubicin lipo-


somes, deploying therapeutically at 39° to 42°C.
This favorable thermal profile may facilitate Bilayer crosslink
drug deposition, allowing for more drug to
deploy where needed with less systemic toxic-
ity (29).
A nanoparticle that targets angiogenesis
Bound metal tor imaging --=-..........
recently has been developed that uses the Clv ~3
endothelial cell surface integrin as a target (Fig. FIGURE 22.13. Nanoparticle delivered in the blood-
22.13). This integrin is upregulated and strongly stream with specific tumor targeting may be an ideal
expressed in tumor vessel endothelium, and combination treatment for image-guided electro-
may be better than vascular endothelial growth magnetic energy therapy like RFA.
factor (VEGF). Polyvalency also may allow
more specific targeting. This nanoparticle also increased permeability effect in the sublethally
can kill tumors by delivering genes selectively heated residual tumor. Many pharmaceutical
to growing blood vessels on tumor-bearing treatments will be combined with thermal abla-
mice (30). Expression of this integrin may tion to improve pharmacokinetics and efficacy
be imaged by PET or MR via a monoclonal (Fig. 22.10).
antibody attached to paramagnetic contrast.
Molecular imaging may provide a surrogate Conclusion
marker for incomplete tumor ablation, thus
facilitating follow-up, prognosis, response, and One can envision the day when the treatment
early interventions before becoming geo- plan will be constructed by a computer program
metrically unfavorable for repeat treatment. that decides where the best entry point is and
However, the effects of heat on angiogenesis the best needle position to avoid heat sink. The
are likely variable and complex. RFA treatment planning program also will
Each method of targeting may allow for a calculate thermal dosimetry similar to radiation
higher order of specificity, so heat activation therapy planning. It will tell you whether
may augment local delivery rationales for many balloon occlusion or the Pringle maneuver is
engineered drugs with molecular targeting. required. The entry points and targets will be
Defining the genomics and proteomics of tumor clicked with a mouse, and electromagnetic
heating may classify upregulated genes that focusing needles will be placed by a robot,
could be used to improve heat-mediated cell with physician assistance. Magnetic tracking
targeting or local gene delivery or transfection. within the needle will provide navigation
Selective local/regional drug delivery also using multimodality image fusion for deploying
will become more relevant in the future as a predetermined treatment plan based on
patient-specific molecular targets are identified. patient-specific anatomic and perfusion data
Definition of temporal and spatial hetero- with a dielectric feedback loop to optimize the
geneities of specific tumors will lead to tissue-specific treatment algorithm. The organ-
patient-specific drug cocktails to be adminis- specific module will be plugged into the gener-
tered at specific times, according to molecular ator and the tissue will be cooked, at which
imaging-defined guidelines. Radiofrequency point image fusion will automatically identify
ablation has the attractive features of debulk- areas of residual tumor. Augmented reality will
ing part of the tumor burden, and simul- display the anatomy below the skin when the
taneously increasing drug deposition via an needle is pointed toward the target before
22. New Technologies in Tumor Ablation 299

the needle is physically placed. Needle-based 10. Kampinga HH, Dikomey E. Hyperther-
sensors will use optical spectroscopy to charac- mic radiosensitization: mode of action and
terize chromatin density and identify tumor clinical relevance. Int J Radiat Bioi 2001;77:399-
margins. An intravenous or local intraarterial 408.
11. Mir LM, Rubinsky B. 'Iteatment of cancer with
injection of a molecularly targeted liposome
cryochemotherapy. Br J Cancer 2002;86:1658-
will be performed to turn local RFA into
1660.
regional or systemic therapy. Radiofrequency 12. Gao J, Qian F, Szymanski-Exner A, Stowe N,
ablation, electroporation, or sonoporation will Haaga 1. In vivo drug distribution dynamics in
increase therapeutic gene transfection of the thermoablated and normal rabbit livers from
liposome payload. The physician pilot will be biodegradable polymers. J Biomed Mater Res
drinking coffee in the control room to make 2002;62:308-314.
sure he stays awake. 13. Weaver JC, Chizmadzhev YA. Theory of elec-
troporation: a review. Bioelectrochem Bioener-
References getics 1996;41:135-160.
14. Liu F, Huang L. Improving plasmid DNA-
1. Thngjitkusolmun S, Staelin ST, Haemmerich D, mediated liver gene transfer by prolonging its
et al. Three-dimensional finite-element analyses retention in the hepatic vasculature. J Gene Med
for radio-frequency hepatic tumor ablation. 2001;3:569-576.
IEEE Trans Biomed Eng 2002;49:3-9. 15. Mir LM, Glass LF, Sersa G, et al. Effective treat-
2. Solomon SB, Patriciu A, Bohlman ME, Kavoussi ment of cutaneous and subcutaneous malignant
LR, Stoianovici D. Robotically driven interven- tumours by electrochemotherapy. Br J Cancer
tions: a method of using CT fluoroscopy without 1998;77:2336-2342.
radiation exposure to the physician. Radiology 16. Sersa G, Stabuc B, Cemazar M, Miklavcic D,
2002;225:277-282. Rudolf Z. Electrochemotherapy with cisplatin:
3. Bloom MB, Salzberg AD, Krummel TM. clinical experience in malignant melanoma
Advanced technology in surgery. Curr Probl patients. Clin Cancer Res 2000;6:863-867.
Surg 2002;39:733-830. 17. Cemazar M, Sersa G, Wilson J, et al. Effective
4. Su LM, Stoianovici D, Jarrett TW, et al. Robotic gene transfer to solid tumors using different non-
percutaneous access to the kidney: comparison viral gene delivery techniques: electroporation,
with standard manual access. J Endourol 2002; liposomes, and integrin-targeted vector. Cancer
16:471-475. Gene Ther 2002;9:399-406.
5. Wood BJ, Banovac F, Friedman M, et al. CT- 18. Davalos RV, Rubinsky B, Otten DM. A feasibil-
integrated programmable robot for image- ity study for electrical impedance tomography
guided procedures: comparison of free-hand and as a means to monitor tissue electroporation for
robot-assisted techniques. J Vasc Intervent molecular medicine. IEEE Trans Biomed Eng
RadioI2003;14:S62. 2002;49:400-403.
6. Vekris A, Maurange C, Moonen C, et al. Control 19. Miller DL, Quddus 1. Sonoporation of mono-
of transgene expression using local hyperther- layer cells by diagnostic ultrasound activation of
mia in combination with a heat-sensitive pro- contrast-agent gas bodies. Ultrasound Med BioI
moter. J Gene Med 2000;2:89-96. 2000;26:661--667.
7. Kruskal JB, Goldberg SN. Emerging therapies 20. Ward M, Wu J, Chiu JE Experimental study of
for hepatocellular carcinoma: opportunities for the effects of Optison concentration on sonopo-
radiologists. J Vase Intervent Radiol 2002;13: ration in vitro. Ultrasound Med BioI 2000;26:
S253-S258. 1169-1175.
8. Schratzberger P, Krainin JG, Schratzberger G, 21. Wu J, Ross JP, Chiu JE Reparable sonoporation
et al. Transcutaneous ultrasound augments generated by microstreaming. J Acoust Soc Am
naked DNA transfection of skeletal muscle. Mol 2002;111:1460-1464.
Ther 2002;6:576--583. 22. Wu 1. Theoretical study on shear stress generated
9. Zhang J, Dong B, Liang P, et al. Significance by microstreaming surrounding contrast agents
of changes in local immunity in patients with attached to living cells. Ultrasound Med Bioi
hepatocellular carcinoma after percutaneous 2002;28:125-129.
microwave coagulation therapy. Chin Med J 23. Anwer K, Kao G, Proctor B, et al. Ultrasound
(Engl) 2002;115:1367-1371. enhancement of cationic lipid-mediated gene
300 B.J. Wood et al

transfer to primary tumors following systemic 27. Bednarski MD, Lee JW, Callstrom MR, Li KC.
administration. Gene Ther 2000;7:1833-1839. In vivo target-specific delivery of macromolecu-
24. Husseini GA, Runyan CM, Pitt WG. Investi- lar agents with MR-guided focused ultrasound.
gating the mechanism of acoustically activated Radiology 1997;204:263-268.
uptake of drugs from Pluronic micelles. BMC 28. Goldberg SN, Kamel IR, Kruskal JB, et a1.
Cancer 2002;2:20. Radiofrequency ablation of hepatic tumors:
25. Gnant MF, Noll LA, Terrill RE, et a1. Isolated increased tumor destruction with adjuvant lipo-
hepatic perfusion for lapine liver metastases: somal doxorubicin therapy. AJR 2002;179:93-
impact of hyperthermia on permeability of 101.
tumor neovasculature. Surgery 1999;126:890- 29. Kong G, Braun RD, Dewhirst MW. Characteri-
899. zation of the effect of hyperthermia on nano-
26. Kruskal JB, Oliver B, Huertas JC, Goldberg particle extravasation from tumor vasculature.
SN. Dynamic intrahepatic flow and cellular alter- Cancer Res 2001;61:3027-3032.
ations during radiofrequency ablation of liver 30. Hood JD, Bednarski M, Frausto R, et al. Tumor
tissue in mice. J Vasc Intervent Radiol 2001; regression by targeted gene delivery to the neo-
12:1193-1201. vasculature. Science 2002;296:2404-2407.
23
Combination Therapy for Ablation
Allison Gillams and William R. Lees

Radiofrequency ablation (RFA) is most effec- immediately adjacent to the area of ablation in
tive for small tumors. In metastases, complete a cohort of 32 patients (7). Tissue perfusion-
treatment has been reported in 91 % of tumors mediated cooling, or the "heat-sink effect,"
<2 cm, 88% of tumors <3 cm, and 53 % of 3 to restricts the volume of ablation that can be
4.5 cm tumors (1,2). In hepatocellular carci- achieved. The relationship between tissue
noma (HCC), RFA achieved complete necrosis perfusion and the volume of necrosis has been
in 90% of lesions <3cm and 71 % of lesions 3.1 elegantly demonstrated using pharmacologic
to 5.0cm in diameter (3). In our practice, most manipulation. For example, a halothane-
patients present with larger lesions. induced 46% reduction in blood flow resulted
Many patients develop new disease following in a doubling of the diameter of the area of
successful ablation of colorectal metastases; necrosis (8). Further experiments with occlu-
50% develop new metastases elsewhere in the sion of the hepatic artery, portal vein, or both
liver (4) and 40% develop extrahepatic disease vessels in animals produced increased amounts
(5). For HCC, the 5-year recurrence rate in the of necrosis (9). In one study, hepatic arterial
liver following successful resection varies from occlusion increased the area of necrosis 2.5-
67.6% to 100% (6). The vast majority of these fold, portal venous occlusion by 1.5 times, and
patients eventually succumb to their disease. occlusion of both vessels by 5.7-fold (10). In a
Therefore, combination therapies are required second study, using a different RFA system, the
not only to increase the local ablative efficacy volume of necrosis was increased 1.8-fold by
for larger lesions, but also to reduce the inci- hepatic arterial occlusion, twofold by portal
dence of new lesions and extrahepatic disease. venous occlusion, and 2.9-fold by occlusion of
both vessels (11).
Balloon occlusion is practiced in some
Combination Therapies to centers, particularly for the treatment of large
HCCs (12). At surgery, the vascular pedicle can
Improve Local Ablation Efficacy be clamped. It must be remembered that while
vascular occlusion increases the volume of
Vascular Occlusion and
necrosis, it also removes the protective effect
Radiofrequency Ablation
of cooling blood, and there is a predictable
Normal liver, like other tissues, responds to increase in bile duct injury (10). This could be
thermal injury with an increase in arterial important for treatment adjacent to the main
perfusion. This change has been quantified bile ducts.
using contrast-enhanced computed tomogra- Another option for patients undergoing treat-
phy (CT). A mean 3.2-fold increase in hepatic ment under general anesthesia is "hypotensive
arterial perfusion was observed in normal liver anesthesia". This technique, employed in liver

301
302 A. Gillams and w.R. Lees

resection, is the controlled reduction of the the laparoscopic approach could combine the
systolic blood pressure to approximately 80 advantage of reduced invasiveness offered by
mmHg. In one study, this maneuver resulted in the percutaneous approach and better staging
a 44% increase in necrosis volume (13). inherent in the surgical approach. However, the
use of laparoscopic US to guide treatment is
difficult and requires substantial skill. A com-
Surgery and Radiofrequency Ablation parison of open and laparoscopic approaches
for ablation in an animal model using 1-cm
At Open Laparotomy
target lesions showed a higher positive margin
Radiofrequency ablation can be performed in rate, 33%, with the laparoscopic approach vs.
conjunction with surgical resection (14). In 9% with the open approach (18).
some patients, the size and distribution of their
metastases make this the optimal approach.
Radiofrequency ablation can be used to Intratumoral Injection and
increase the number of patients who are eligi- Radiofrequency Ablation
ble for an ablative procedure. Advantages of Ethanol
the open surgical approach include better
staging and improved detection of very small Injection of 100% ethanol into the tumor has
liver metastases with intraoperative ultrasound been used in conjunction with thermal ablation
(US). Disadvantages of this approach include to increase ablation efficacy. The postulated
the invasiveness, complication rate, expense, mechanism is vasoconstriction of tumor vessels
and the lack of CT and magnetic resonance that reduces the heat-sink effect. Our study in
imaging (MRI) monitoring of the procedure. 19 patients treated with laser demonstrated a
Intraoperative RFA, therefore, is indicated only small (mean 5.4%) increase in the volume of
when another procedure is required, for ex- necrosis.
ample, liver or bowel resection. Cryotherapy
has been used to successfully reduce the inci- Saline
dence of edge recurrence following resection Several groups have explored the use of saline
in patients with inadequately treated margins; to increase the volume of necrosis (19). The
RFA could be used in a similar fashion (15). It saline is thought to act as a wet electrode by
has also been used to produce a zone of dessi- increasing the effective electrode surface and
cation at the resection margin with a reported hence the flux of RF current. The effect
reduction in blood loss (16). increases with the concentration of saline (20).
Saline infusion can be performed as an adjunc-
Laparoscopic Approach tive technique, and also has been incorporated
into electrode design. Ex vivo and in vivo
Laparoscopic RFA is useful when contiguous experiments with "wet electrodes" have shown
structures that may be damaged by the ablation larger areas of necrosis, but also a more irregu-
need to be moved away from the area to be lar necrosis (21).
treated. This is particularly important for
bowel, stomach, or diaphragm. Some centers Hepatocellular Carcinoma vs.
use percutaneous injection of sterile water both
Liver Metastases
to separate important structures from the abla-
tion area and to improve ultrasound visualiza- Intratumoral injection usually is more effective
tion of the diaphragmatic surface of the liver. in HCC in which the injectate remains localized
Laparoscopic RFA has been recommended for within a well-defined lesion. Injection into scir-
HCCs that are poorly visualized transcuta- rhous metastases can be difficult, and extrava-
neously, or for large HCCs that require multi- sation of the injectate often is seen on US. We
ple electrode placements that confer the no longer use saline infusion on grounds of
potential for hemorrhage (17). Theoretically, safety.
23. Combination Therapy for Ablation 303

Radiofrequency Ablation and suppression with attendant thrombocytopenia


and neutropenia. Central lines, in place for the
Systemic Therapies administration of chemotherapy, are a common
source of bacteremia and secondary infection
Chemotherapy of the necrotic ablation area in the liver, and are
Systemic Chemotherapy for Colorectal associated with a high morbidity. As a result of
Liver Metastases this additional comorbidity, we have tended to
avoid concurrent systemic chemotherapy; we
There have been exciting developments in the prefer to use chemotherapy prior to RFA to
treatment of colorectal liver metastases with downstage patients with more extensive disease
chemotherapy. For the first time, a significant, than is amenable to RFA alone.
if small, improvement in survival has been
achieved with irinotecan (22). Another new
regime, Oxaliplatin and 5-fluorouracil (5-FU), Chemoembolization and
has been shown to have higher response rates, Radiofrequency Ablation for
as much as 53%, compared to 5-FU and folinic Liver Metastases
acid, in which the response rate was 28% (23).
Trials are in progress evaluating these new Although chemoembolization is not a well-
agents alone and in conjunction with 5-FU, as accepted therapy for metastatic disease, there
first- or second-line therapy. are several reports that show response rates
varying between 25% and 100%, with median
survival between 7 and 23 months (29). The
Neoadjuvant Chemotherapy response is transient, but if a reduction in size
Surgical resection has been performed effec- is achieved, then it is feasible that this could be
tively in patients who have been downstaged by followed by RFA. We are currently exploring
chemotherapy (24,25). Similarly, our results the role of chemoembolization followed by
with RF have shown that chemoresponders can RFA for larger (>5cm) metastases (Fig. 23.1).
benefit from ablation (26). The survival from
the diagnosis of liver metastases in chemore-
sponders was 39 months, compared to 42 Systemic Chemotherapy in Breast
months for patients who had either not Liver Metastases
received chemotherapy or had not been down- Radiofrequency ablation has been used suc-
staged by it. With more efficacious first-line cessfully in patients with limited liver metas-
agents becoming available, many more patients tases (30). Some of these patients have had
will become suitable for ablative procedures, extrahepatic disease, often considered a
either surgical or RFA. contraindication to local ablative therapy.
However, there is evidence that treated bony
Adjuvant Chemotherapy and mediastinal nodal metastases can be stabi-
Radiofrequency ablation does not preclude lized for long periods of time, such that RFA for
chemotherapy as a concurrent treatment. localized liver metastases becomes a reason-
Intraarterial chemotherapy has been used fol- able consideration.
lowing both cryotherapy and in a small group
of patients post-RFA (27,28). In the cryother-
apy group, for those patients who managed to
Neuroendocrine Metastases
complete more than 3 months of chemother- Radiofrequency ablation, cryotherapy, and
apy, the median survival was 582 days com- surgery have been used to increase the effi-
pared to 331 days for those treated with cacy of somatostatin analogues in patients
cryotherapy alone. For patients on systemic who have developed uncontrolled hormone-
chemotherapy, RFA treatment needs to be secretion-related symptoms, despite optimal
scheduled to avoid periods of bone marrow medical therapy (31).
304 A. Gillams and W.R. Lees

A B

Figure 23.1. (A) Computed tomograpy (CT)


scan demonstrates a large solitary hepatocellular
carcinoma. This was treated with a combination of
chemoembolization and radiofrequency ablation.
(B) CT scan postchemoembolization showing good
Lipiodol uptake posteriorly, but poor uptake anteri-
orly. There is evidence of necrosis centrally, but resid-
ual tumor around the periphery. Four weeks after
embolization, radiofrequency ablation was per-
formed with a triple water-cooled electrode. (C)
c Follow-up CT scan shows a large area of necrosis.

Radiotherapy/Chemotherapy and time of treatment. Undoubtedly there will be a


Radiofrequency Ablation-Induced role for this type of therapy, but the exact role
Hyperthermia is still to be defined.
Radiofrequency ablation requires tempera-
tures in excess of 55°C to produce cell death.
Lower temperatures, 42° to 46°C, so-called ChemotherapyIPrevention in
hyperthermia, result in reversible cell damage,
but if used in conjunction with radiotherapy or
Hepatocellular Carcinoma
chemotherapy will produce necrosis. Animal
Combination Transcatheter Arterial
experiments in a rat breast tumor model have
Chemoembolization /Ablation
demonstrated an improved volume of necrosis
when RFA was used in conjunction with Transcatheter arterial chemoembolization
chemotherapy (32). In an early clinical study in (TACE) alone results in complete necrosis in
10 patients with various liver tumors, a single only 38% of lesions <3cm in diameter (33). It
bolus of IV liposomal doxorubicin 24 hours has been combined with percutaneous ethanol
prior to RFA resulted in good areas of necro- injection (PEl) and has been shown to be supe-
sis with extension of the necrotic areas over the rior to PEl alone, in reducing both the inci-
ensuing 30 days. The downside of this approach dence of local residual disease as well as the
is the inability to predict the final result at the number of new foci of HCC that arise in
23. Combination Therapy for Ablation 305

previously normal liver (34). Survival was not Conclusion


affected.
Radiofrequency ablation is effective in treat- While the development of thermal ablation
ing HCC up to 4.5 cm. However, in the absence represents an exciting advance in the treatment
of screening programs, many patients present of liver cancer, it is likely that a combination
with larger lesions. Chemoembolization has of different therapeutic modalities will be
been used in conjunction with thermal ablation, required for most patients.
both laser and RF, for large (5-8cm) HCCs.
Different techniques have been implemented,
for example, laser ablation followed by TACE
after an interval of 30 to 90 days, or RFA with References
balloon occlusion, followed immediately by
1. Solbiati L, Livraghi T, Goldberg SN, et al. Per-
selective embolization (35,36). Using laser fol- cutaneous radiofrequency ablation of hepatic
lowed by TACE, Pacella et al (35) reported a metastases from colorectal cancer: long-term
90% rate of complete necrosis in large (mean results in 117 patients. Radiology 2001;221:159-
5.2cm) HCCs. 166.
2. de Baere T, Elias D, Dromain C, et al. Radio-
frequency ablation of 100 hepatic metastases
Prevention with a mean follow-up of more than 1year. AJR
Several agents have been explored to attempt 2000;175:1619-1625.
to reduce the incidence of HCC in high-risk 3. Livraghi T, Goldberg SN, Lazzaroni S, et al.
patients. Immunopreventions, such as inter- Hepatocellular carcinoma: radiofrequency
ablation of medium and large lesions. Radiology
feron and glycyrrhizin, have been used to
2000;214:761-768.
suppress the development of HCC. Interferon 4. Gillams AR, Lees WR. Radiofrequency ablation
prevents HCC in two groups-in those patients of colorectal metastases in 167 patients. Eur
with long-term elimination of the hepatitis C Radio12004;14(12):2261-2267.
virus (15-35% of patients), and in those with 5. Chopra S, Dodd GD III, Chintapalli KN, et al.
sustained normalization of serum transami- Tumor recurrence after radiofrequency thermal
nases (an additional 10%). Interferon has ablation of hepatic tumors: spectrum of findings
been shown to reduce recurrence after surgery on dual-phase contrast-enhanced CT. AJR 2001;
(37). Glycyrrhizin, on the other hand, has no 177:381-387.
antiviral activity, but acts by reducing liver 6. Izumi N,Asahina Y, Noguchi 0, et al. Risk factors
inflammation as indicated by suppressed for distant recurrence of hepatocellular carci-
transaminases (38). noma in the liver after complete coagulation by
microwave or radiofrequency ablation. Cancer
The chemopreventive agent polyprenoic acid 2001;91:949-956.
an acyclic retinoid, induces the disappearance 7. Gillams AR, Lees WR. Thermal ablation
of serum lectin reactive a-fetoprotein, which induced changes in hepatic arterial perfusion.
suggests a clonal deletion of minute premalig- Radiology 1999;213P;382.
nant cells. It has been shown to reduce the 8. Goldberg S, Hahn P, Halpern E, Fogle R,
incidence of recurrent or new HCC postcu- Gazelle GS. Radiofrequency tissue ablation:
rative resection 27% vs. 49% at 38 months effect of pharmacological modulation of blood
(39). flow on coagulation diameter. Radiology 1998;
Finally, another technique that had more 209:761-767.
modest success in reducing the incidence of 9. Germer CT, Isbert C, Albrecht D, et al. Laser-
induced thermotherapy combined with hepatic
recurrence postresection was a single dose of
arterial embolization in the treatment of liver
intraarterial 1311 Lipiodol. This resulted in a tumors in a rat tumor model. Ann Surg 1999;
recurrence rate of 28.5% vs. 59% at a median 230:55-62.
follow-up of 34.6 months (40). In the future, a 10. Denys A, de Baere T, Mahe C, et al. Radiofre-
combination of immuno- and chemopreven- quency tissue ablation of the liver: effects of
tions will be explored. vascular occlusion on lesion diameter and biliary
306 A. Gillams and WR. Lees

and portal damages in a pig model. Eur Radiol 23. Giacchetti S, Perpoint B, Zidani R, et al. Phase
2001;11:2102-2108. III multicenter trial of oxaliplatin added to
11. Chinn SB, Lee FT, Kennedy GD, et al. Effect of chronomodulated fluorouracil-leucovorin as first
vascular occlusion on radiofrequency ablation line treatment of metastatic colorectal cancer.
ofthe liver: results in a porcine model.AJR 2001; J Clin OncoI2000;18:136-147.
176;789-795. 24. Bismuth H, Adam R, Levi F, et al. Resection of
12. Yamasaki T, Kurokawa F, Shirahashi H, et al. nonresectable liver metastases from colorectal
Percutaneous radiofrequency ablation therapy cancer after neoadjuvant chemotherapy. Ann
with combined angiography and CT assistance Surg 1996;224:509-522.
for patients with hepatocellular carcinoma. 25. Shankar A, Leonard P, Renaut AJ, et al. Neoad-
Cancer 2001;91:1342-1348. juvant therapy improves resectability rates for
13. Lees WR, Schumillian C, Gillams AR. Hypoten- colorectal cancer. Ann R ColI Surg Engl2oo1 ;83:
sive anesthesia improves the effectiveness of 85-88.
radiofrequency ablation in the liver. Radiology 26. Gillams A, Lees W Should thermal ablation be
2000;217:228. offered to patients with colorectal liver metas-
14. Wood TF, Rose DM, Chung M, Allegra Dp, tases downstaged by chemotherapy? Radiology
Foshag U, Bilchik Al Radiofrequency ablation 2000;217(P):539.
of 231 unresectable hepatic tumors: indications, 27. Preketes A, Caplehorn J, King J, et al. Effect
limitations and complications. Ann Surg Oncol of hepatic artery chemotherapy on survival of
2000;7:593-600. patients with hepatic metastases from colorectal
15. Gruenberger T, Jourdan JL, Zhao J, King J, carcinoma treated with cryotherapy. World J
Morris DL. Reduction in recurrence risk for Surg 1995;19:768-771.
involved or inadequate margins with edge 28. Kainuma 0, Asano T, Aoyama H, et al. Com-
cryotherapy after liver resection for colorectal bined therapy with radiofrequency thermal
metastases. Arch Surg 2001;136:1154-1157. ablation and intra-arterial infusion chemother-
16. Weber JC, Navarra G, Jiao LR, Nicholls Jp, apy for hepatic metastases from colorectal
Jensen SL, Habib I. New technique for liver cancer. Hepatogastroenterology 1999;46:1071-
resection using heat coagulation necrosis. Ann 1077.
Surg 2002;236(5):560-563. 29. Tellez C, Benson A, Lyster M, et al. Phase II trial
17. Montorsi M, Santambrogio R, Bianchi P, et al. of chemoembolization for the treatment of
Radiofrequency interstitial thermal ablation of metastatic colorectal carcinoma to the liver and
hepatocellular carcinoma in liver cirrhosis: role review of the literature. Cancer 1998;82:1250-
of the laparoscopic approach. Surg Endosc 2001; 1259.
15:141-145. 30. Livraghi T, Goldberg N, Solbiati L, et al. Percu-
18. Scott DJ, Fleming JB, Watumull LM, et al. taneous radiofrequency ablation of liver metas-
Accuracy and effectiveness of laparoscopic vs. tases from breast cancer: initial experience in 24
open hepatic RF ablation. Surg Endosc 2001;15: patients. Radiology 2001;220:145-149.
135-140. 31. Chung MH, Pisegna J, Spirt M, et al. Hepatic
19. Miao Y, Ni Y, Yu J, Marchal G. A comparative cytoreduction followed by a novel long-acting
study on validation of a novel cooled-wet elec- somatostatin analogue: a paradigm for in-
trode for radiofrequency liver ablation. Invest tractable neuroendocrine tumors metastatic to
Radiol 2000;35:438-444. the liver. Surgery 2001;130:954-964.
20. Goldberg SN, Ahmed M, Gazelle GS, et al. 32. Goldberg SN, Saldinger P, Gazelle GS, et al. Per-
Radio-frequency thermal ablation with NaCl cutaneous tumor ablation: increased necrosis
solution injection: effect of electrical conductiv- with combined radiofrequency ablation and
ity on tissue heating and coagulation-phantom intratumoral doxorubicin in a rat breast tumor
and porcine liver study. Radiology 2001;219: model. Radiology 2001;220:420-427.
157-165. 33. Higuchi T, Kikuchi M, Okazaki M. Hepatocellu-
21. Livraghi T, Goldberg N, Monti F, et al. Saline lar carcinoma after transcatheter hepatic arterial
enhanced radiofrequency tissue ablation in the embolization. A histopathological study of 84
treatment of liver metastases. Radiology 1997; resected cases. Cancer 1994;72:2259-2267.
202:205-210. 34. Koda M, Murawaki Y, Mitsuda A, et al.
22. Douillard JY, V303 Study Group. Irinotecan and Combination therapy with transcatheter arterial
high dose fluorouracil/leucovorin for metastatic chemoembolization and percutaneous ethanol
colorectal cancer. Oncology 2000;14:51-55. injection compared with percutaneous
23. Combination Therapy for Ablation 307

ethanol injection alone for patients with small after complete resection or ablation of the
hepatocellular carcinoma. Cancer 2001;92:1516- primary tumor. Hepatology 2000;32:228-232.
1524. 38. Okuno M, Kojima S, Moriwaki H. Chemopre-
35. Pacella CM, Bizzarri G, Cecconi 0, et al. Hepa- vention of hepatocellular carcinoma: concept
tocellular carcinoma: long-term results of com- progress and perspectives. J Gastroenterol
bined treatment with laser thermal ablation Hepatol2001;16:1329-1355.
and transcatheter arterial chemoembolization. 39. Muto Y, Moriwaki H, Ninomiya M, et al. Pre-
Radiology 2001 ;219:669-678. vention of second primary tumors by an acyclic
36. Lencioni R, Cioni D, Donati P, Bartolozzi C. retinoid, polyprenoic acid in patients with HCC.
Combination of interventional therapies in N Engl J Med 1996;334:1561-1567.
hepatocellular carcinoma. Hepatogastroenterol- 40. Lau WY, Leung TWT, Ho S, et al. Adjuvant
ogy 2001;48:8-14. intra-arterial iodine-131 labelled lipiodol for
37. Ikeda K, Arase Y, Saitoh S, et al. Interferon beta resectable hepatocellular carcinoma: a prospec-
prevents recurrence of hepatocellular carcinoma tive randomized trial. Lancet 1999;353:797-801.
Section V
Organ System Tumor Ablation
24
Ablation of Liver Metastases
Luigi Solbiati, Tiziana Ierace, Massimo Tonolini, and Luca Cova

Metastatic liver disease represents one of the series this technique demonstrated significant
most common clinical problems in oncology advantages including (a) feasibility of treat-
practice. MUltiple treatment options are ment in previously resected patients and non-
available including hepatic resection, chemo- surgical candidates due to extent of metastatic
embolization, intraarterial and systemic chemo- involvement, age, and comorbidity; (b) repeata-
therapy, cryotherapy, and radiofrequency bility of treatment when incomplete and when
ablation (RFA) (1,2). local recurrence or development of metach-
Over the past few years, advances in diag- ronous lesions occur; (c) combination with sys-
nostic imaging modalities such as contrast- temic or regional chemotherapy; (d) minimal
enhanced ultrasound, single- and multidetector invasiveness with limited complications rate
helical computed tomography (CT) and mag- and preservation of liver function; (e) limited
netic resonance imaging (MRI) with hepatobil- hospital stays and procedure costs (29-42).
iary and reticuloendothelial-specific contrast
agents have achieved early detection and
accurate quantification of hepatic metastatic
involvement (3-10). As a result, correct selec- Pathology of Metastatic
tion of patients for different treatment options Liver Disease
is usually possible.
When feasible, surgical resection of hepatic After regional lymph nodes, the liver is the
metastases is the accepted standard therapeu- organ most commonly involved with metastatic
tic approach in patients with colorectal cancer. disease, usually from primary cancers of the
Surgery also offers potential for cure in selected gastrointestinal tract, breast, and lung. The pres-
patients with other primary tumors (2,11-28). ence of a dual blood supply (from the arterial
Percutaneous radiofrequency thermal abla- system and the portal/splanchnic venous
tion is an established therapeutic option for system) and the discontinuous endothelium of
liver metastases, which may obviate the need the hepatic sinusoids (allowing communication
for a major surgery and result in prolonged sur- with the extracellular space) account for the
vival and chance for cure. Extensive operators' high incidence of metastatic spread to the liver.
experience and technical advances provide Morphologic features of liver metastases
larger coagulation volumes and therefore allow as well as their localization and number may
safe and effective treatment of medium- and be extremely variable on gross pathology.
even large-size metastases. Results of RFA in Histopathology explains the lack of the pecu-
terms of global and disease-free survival cur- liar "oven effect" observed and understood
rently approach those reported for surgical during experience with RFA of hepatocellular
metastasectomy. Furthermore, in published carcinoma (HCC). During treatment of HCC,

311
312 L. Solbiati et al

the tumor capsule and the surrounding fibrotic, lll]ection of doxorubicin in liposome carriers
poorly vascularized cirrhotic parenchyma before the procedure (49).
prevent thermal conduction outside the treated
lesion; therefore, optimal heat diffusion is main-
tained in the softer, usually well-circumscribed Patient Selection and Indications
tumoral nodule (34). Liver metastases are for Treatment
not encapsulated and tend to infiltrate the
surrounding well-vascularized liver tissue that In the early phase of our experience with RFA,
limits tumor heating (32,33). treatment was performed in patients with new
Analysis of surgical series demonstrated that metastases or local recurrences after previous
after resection of liver metastases, local recur- hepatic resection and in patients refusing or not
rence is frequent if a surgical margin of 1em of eligible for surgery for technical or general
normal hepatic tissue surrounding the tumor is health reasons. Currently, increasing awareness
not removed (15). Similarly, RFA of liver by referring oncologists of the reported results
metastases should aim to ablate a 0.5- to 1.0- and of the minimal invasiveness of RFA
em-thick "safety margin" of peritumoral liver make this technique a valid therapeutic option
tissue to kill infiltrating tumor undetectable by for local treatment of patients with limited
currently available imaging modalities, thus metastatic liver disease.
reducing the risk of recurrence (30). Radiofrequency ablation is applicable to
Therefore, adequate RFA treatment of liver patients with one to five metachronous liver
metastases often requires the creation of large metastases up to 3.5 to 4cm in largest diameter
volumes of coagulation necrosis. Recent tech- from previous radically treated colorectal or
nical innovations including internally cooled other primary cancer, in whom surgery is not
electrodes (43), cluster electrodes (44), pulsed feasible, contraindicated, or not accepted. Some
RF energy application (45), and peritumoral patients with large lesions or more than five
saline injection prior to or during energy depo- nodules may undergo RFA after successful
sition (46) enable better energy deposition and tumor debulking by means of chemotherapy
increase coagulation necrosis volumes, thus (Table 24.1).
allowing extended indications for RFA. We consider the presence of active extrahe-
More recently, further improvements in patic disease a contraindication to RF treat-
induced coagulation have been achieved. These ment, with the exception of bone or lung
include decreasing blood flow during ablation metastases in breast cancer patients if they
using balloon occlusion of a hepatic vein or seg- are responding or unchanged with systemic
mental portal branch (47), by adopting strate- chemotherapy (30). Exclusion criteria include
gies of precise overlapping of subsequent the presence of severe coagulopathy, renal
electrode placements (48), or by adjuvant IV failure, or jaundice. Informed consent of the

TABLE 24.1. Indications for radiofrequency (RF) ablation treatment of patients with liver metastases.
Primary tumor (most frequently colorectal or breast cancer) previously treated radically with surgery, radiotherapy, and
without adjuvant chemotherapy
No evidence of active extrahepatic disease; possible exception for breast cancer metastases in the lung or bone showing
regression or stability over time on hormonal or chemotherapy
Liver metastasis (cyto/histologically confirmed, compatible with primary tumor)
Residual tumor after previous treatment with RF ablation or other treatment
Local recurrence after surgical resection, RF ablation. or other treatment
Metachronous new metastasis after previous resection, RF ablation, or other treatment
Up to 4 to 5 lesions. each 4cm or less in maximum diameter; lesions over 4cm may be treated after debulking by
systemic or regional chemotherapy
Metastases visible with B-mode and/or contrast-enhanced ultrasound
Feasible and safe percutaneous access (not abutting the hepatic hilum, the gallbladder, or the colon)
Adequate coagulation, hepatic and renal function
Informed consent obtained
24. Ablation of Liver Metastases 313

patient is obtained after explanation of the pro- trast agents may allow even higher sensitivity
cedure, the expected results, and the possible rates (3).
complications.
Accurate assessment of the extent of
metastatic liver disease is crucial to optimal Procedure
patient selection. Intraoperative ultrasound
(IOUS) is still considered the "gold standard" In OUr department RFA treatment sessions
for detection of focal liver lesions. At OUr insti- are carried out in a dedicated operating rOom
tution, candidates for RFA of hepatic metas- under general anesthesia using endotracheal
tases undergo pretreatment diagnostic workup intubation and mechanical ventilation. Some
that includes laboratory tests, tumor markers, other centers adopt conscious sedation.
conventional and contrast-enhanced liver ultra- A 200-W, 480-kHz RF generator system
sound, and single- Or multidetector helical CT (Radionics, Burlington, MA) with an auto-
examinations. Tumor staging usually is com- mated pulsed-RF algorithm is used together
pleted by thoracic CT and radionuclide bone with single or cluster internally cooled elec-
scan to exclude distant tumor dissemination. trodes. Usually percutaneous RFA is per-
Whereas ultrasound is the most commonly formed under contrast-enhanced ultrasound
used imaging modality in screening for metasta- guidance. In some patients CT guidance is pre-
tic spread, the sensitivity of unenhanced (B- ferred when sonographic targeting is not possi-
mode) ultrasound is significantly affected by the ble, with significantly increased procedure time.
operator's experience as well as the patient's The choice of the electrodes is based on the size
body habitus and presence of bowel gas disten- of the target nodule: for lesions smaller than
tion. Sonography is further limited in enlarged, 2 cm, a single insertion of a 3-cm exposed tip
grossly hyperechoic livers due to steatosis, fibro- single electrode is sufficient. Lesions 2 to 3 cm
sis, chronic liver disease, and postchemothera- in size can be treated with a single insertion of
peutic changes. The detection of isoechoic and a cluster electrode or with multiple insertions
subcentimeter lesions is particularly difficult: in of a 3-cm exposed tip electrode. Lesions ex-
our personal experience a 20% sensitivity rate ceeding 3 cm can be treated only with one to
is not surprising. Contrast-specific ultrasound two insertions of a cluster electrode and one to
software with the use of first-generation (in the two single electrode insertions.
late liver-specific phase) Or second-generation The applied energy is variable, and usually
contrast agents facilitates an increase in detec- reaches 1600 to 1800 rnA for single electrodes
tion rate, particularly for small lesions, as well as and 1800 to 2000 rnA for cluster electrodes.
better characterization of focal lesions (5-9). Each application of energy lasts 12 minutes, and
Precise staging of hepatic neoplastic disease total procedure time ranges from 12 to 15
may require the use of a contrast-enhanced minutes for small solitary lesions to 45 to 60
ultrasound (CEUS) examination to overcome minutes for large or multiple ablations.
limitations of conventional B-mode ultrasound B-mode and color/power Doppler ultra-
and to maximize detection of metastatic lesions, sound are not reliable to assess the size and
possibly modifying the therapeutic approach completeness of induced coagulation necrosis
(8). at the end of the energy application; further-
Multiphase contrast-enhanced helical CT more, additional repositioning of the electrode
currently constitutes the mainstay of hepatic is usually made difficult by the hyperechogenic
imaging, providing overall staging of liver "cloud" that appears around the distal probe.
disease and accurate assessment of resectabil- Therefore, we routinely use contrast-enhanced
ity. Sensitivity of CT was estimated at 85% in ultrasound performed at the presumed end of
surgically controlled series, and missed lesions the treatment, with the patient still under anes-
average 0.7mm in diameter (3,4). In some insti- thesia, to enable rapid assessment of the extent
tutions hepatic staging is obtained by means of of achieved tumor ablation and to discover
dynamic gadolinium-enhanced MRI; the use of viable tumor that requires additional immedi-
reticuloendothelial and liver-specific MR con- ate treatment (50).
314 L. Solbiati et al

Patients receive antibiotic prophylaxis with


ceftriaxone 2 gr prior to the treatment and also
antiemetic and analgesic drugs as needed post-
procedurally. Hospitalization lasts for 48 hours
after the treatment; discharge follows docu-
mentation of necrosis and exclusion of compli-
cations by means of 24-hour contrast-enhanced
CT scan.
In our experience, patients usually experi-
ence mild to moderate pain in the right
abdomen that lasts for 2 to 5 days and is par-
ticularly severe in patients with subcapsular
lesions. Pericapsular and subcapsular injection
of long-acting anesthetic drug (ropivacaine) is
performed at the end of the treatment to
reduce postprocedure pain. Fever may be
present for the first 2 to 3 days after treatment
and recedes with antipyretic drugs (aceta-
minophen). No impairment in liver function
tests has been observed. Morbidity encoun-
tered in a large series of patients treated
with RFA is discussed in the chapter on
complications.
Initial cross-sectional examinations serve to
assess completeness of treatment and provide a
basis for imaging follow-up. However, given the

FIGURE 24.2. A 71-year-old patient with history of


solitary colorectal metastasis at segment VII, treated
with RFA 4 years before. (A) Biphasic CT scan
shows thick, mildly hypodense peripheral rim
located anteriorly to the coagulation area. (B) Local
RFA retreatment has been subsequently performed.

resolution and accuracy of current imaging


techniques, residual microscopic foci of malig-
nancy at the periphery of a treated lesion may
FIGURE 24.1. Contrast-enhanced helical computed sometimes go undetected and give rise to local
tomography (Cf) scan of patient underwent recurrence.
radiofrequency (RF) ablation of two metastases at Contrast-enhanced helical CT and MRI are
segments VII and VIII from colorectal adenocarci- used in the long-term assessment of therapeu-
noma, 9 months before. The area of thermal coagu-
tic response, allowing confident discrimination
lation at segment VII is well demarcated and does
not show signs of local recurrence. Oppositely, along between ablated and residual viable tumor
the medial margin of the area of coagulation at (Figs. 24.1 and 24.2). Cross-sectional imaging
segment VIII, a slightly hypodense oval area due to studies obtained at 3 to 4 months intervals
recurrence is visualized. Furthermore, new tiny are correlated with serum carcinoembryonic
metastases at segments VII, VIII, and II are antigen (CEA) levels to detect local or distant
demonstrated. recurrences. Imaging findings on follow-up
24. Ablation of Liver Metastases 315

examinations with patterns of complete necro- cancer develop recurrent disease that involves
sis and of local recurrences are discussed else- the liver during the course of their diseases.
where in this book. Surgical resection is a potentially curative
In our experience, CEUS has proved valu- treatment for colorectal liver metastases, but
able in the follow-up setting for the detection metastatic liver involvement is often multifocal,
or confirmation of local recurrences and new so that only 20% to 25% of patients have
metastases and for contrast-guided re-treatment resectable disease. Moreover, surgery may be
(48). excluded due to older age and associated
Fluorodeoxyglucose (FDG)-labeled positron pathologic conditions that increase anesthesio-
emission tomography (PET) offers great poten- logic risk, and is associated with prolonged hos-
tial in the evaluation of treatment response pital stays, significant perioperative morbidity,
and is synergistic, and is perhaps superior to and a 2% to 8% mortality rate.
cross-sectional imaging modalities in the sur- Survival rates for resected patients range
veillance of treated patients. Areas of abnormal from 25-28% to 35-40% at 5 years in most
FDG uptake following ablative procedures series, but many successfully resected patients
have been reported to represent disease relapse develop recurrence in the liver and/or extra-
or residual viable tumor with a high degree of hepatic sites, and repeat resection can be per-
sensitivity (50,51). formed in only a minority of them (11-20).
Prognostic factors include stage of primary
tumor, biologic factors (CEA level, differentia-
Combination Therapy tion, ploidy), number of hepatic lesions, size of
dominant lesion, and infiltration of resection
The decision to administer simultaneous margins.
chemotherapy is determined by the referring Regional therapies such as RFA can be
oncologist based on the patient's risk of recur- offered to patients with limited but unresec-
rence, as in patients with poorly differentiated table liver metastases and no extrahepatic dis-
or node-positive primary tumors. Thanks to its ease, whereas chemoembolization and hepatic
minimal invasiveness RFA may be used as a intraarterial chemotherapy are applicable to
first-choice local therapy instead of surgery. patients with extensive liver involvement not
Use of RFA does not prevent the simultaneous suitable for ablation (12,18). Radiofrequency
or subsequent use of other, potentially syner- ablation increases the possibilities of curative
gistic, treatments. treatment in patients with liver recurrence
Hormonal therapy, systemic chemotherapy, after hepatectomy from 17% to 26%, and is
and intraarterial infusional chemotherapy each preferred over repeat surgery because it is less
can be given prior to or after RFA, according invasive (52).
to oncologists' preferences and practice guide- Several published series report promising
lines. If recurrence or development of new results with RFA of liver metastases; however,
lesions is detected, additional RF treatment these early reports had only short-term follow-
sessions may be performed when technically up and included patients with both primary and
feasible after a consensus decision between metastatic liver tumors (37-41).
the radiologist, the oncologist, and the patient. At our institution 166 patients have
been treated with RFA during a 7-year span;
12% had previous surgery for liver neo-
Results: Colorectal Cancer plastic involvement, and repeat resection
was considered unfeasible or too invasive.
Metastases One to five metastases (mean 2.28) were
ablated in each patient for a total of 378 treated
The liver is the first, most common, and often lesions; 64.5% of the treated lesions were
unique site of metastasis of colorectal cancer. 3cm or less in size, and 35.5% were larger than
Approximately 50% of patients with colorectal 3cm.
316 L. Solbiati et al

FIGURE 24.3. A 65-year-old patient with solitary


metachronous colorectal metastasis at segment VII (A).
Since the possibility of surgical resection was excluded
due to cardiovascular disease, single-session RFA was
performed and complete treatment with large coagulation
area was achieved (B). At 3-year follow-up CT scan, no
residual or recurring tumor is found and the necrotic area
has markedly shrunk (C).

B c

In our series, local tumor control was all in 39.1 % of lesions. The incidence of recur-
achieved in 71.4% of lesions (Figs. 24.3 and rence was significantly related to the original
24.4), whereas local recurrence occurred in the size of the treated metastasis: effective local
remaining 28.6% (see Figs. 24.1 and 24.2). tumor control was achieved in 78% of tumors
In the largest published analysis of treated up to 2.5 em, 47% of tumors >2.5 to 4cm, and
patients with colorectal cancer liver metastases 32% of tumors larger than 4 em (36). In another
with follow-up of more than 3 years, including series reported by De Baere and coworkers
our group, local recurrence was observed over- (42), the percutaneous approach allowed local
24. Ablation of Liver Metastases 317

FIGURE 24.4. A 4-year follow-up


CT scan of patient with history of
sigmoid adenocarcinoma and
many metachronous liver metas-
tases, subsequently developing
during the follow-up period. Each
metastasis was treated with cool-
tip RFA. No evidence of residual
or recurrent disease is currently
apparent.

control of 90% of treated lesions after 1 year of of liver metastases to allow additional, still
follow-up, but the mean diameter was signifi- undetected lesions to develop and become
cantly smaller than in our series. The time of identifiable. This limits the number of resec-
recurrence was related to lesion size: local tions carried out in patients who will ultimately
relapse occurred almost always during the first develop more metastases. The use of RFA in
12 months (36). In our patients the median time this setting can decrease the number of resec-
to local recurrence was 6 months. Repeated tions required by obtaining complete tumor
RFA treatment was carried out in 37.6% of control in some patients without major surgery.
metastases with progressive local tumor At the same time, RFA is better accepted by
growth. Including repeat treatments, one to patients than a simple "wait and see" approach
four sessions were performed in each patient, and allows an interval for those who are des-
for a total of 197 sessions (mean 1.18). tined to develop aggressive, disseminated
Distant, metachronous metastases developed disease. In a recent analysis metastasectomy
at follow-up in 36% of our patients. The esti- was spared in 98% of patients with complete
mated median time until detection of new ablation with radiofrequency including 43 % of
metastases was 10 months in our patients, 12
months in the published analysis, and not
significantly related to the original number of
treated lesions (36). 1.00-
In our series, Kaplan-Meier survival analysis
included overall survival rates at 1,2,3,4, and §' 0.75-
5 years of 96.2%, 64.2%, 45.7%, 36.9%, :.cctI
.c
and 22.1 %, respectively; estimated median ~ 0.50-
survival was 33 months (Fig. 24.5). The issue of ~
disease-free survival has been evaluated in the '2::::l 0.25-
cooperative group and rates were calculated as m
49% and 35% at 1 year and 2 years; interest-
0.00-
ingly, time to death was not related to the L o r- - - - - - - - - - - - - - -

number of metastases or to the size of the dom- o 6 12 18 24 30 36 42 48 54 60 66 72 78


survival (months)
inant metastasis (36).
Recently, the "test of time" approach has FIGURE 24.5. The 5-year survival curve of patients
been proposed by surgeons to delay resection treated with RFA for colorectal metastases.
318 L. Solbiati et al

those in whom prior incomplete tumor control were disease-free 4 to 44 months after the treat-
was now achieved by RFA (53). ment. Considering the natural history of the
disease and its minimal invasiveness, RFA is an
effective local therapeutic alternative to sur-
gical resection of liver metastases in patients
Breast Cancer Metastases with breast cancer. Furthermore, RFA may be
extremely useful for tumor recurrences after
Whereas the vast majority of RFA treatments
hepatectomy, since in over half of these patients
address liver metastases from colorectal pri-
the liver is the only involved site of relapse.
maries, a significant subset of secondary hepatic
Combination treatment with hormonal therapy
tumors occurs in patients with breast cancer
or systemic or intraarterial infusion chemother-
(30). Breast cancer, histologically usually ductal
apy usually is required.
adenocarcinoma, is the most frequent neo-
plasm in women and often metastatizes to the
liver.
Metastatic breast cancer traditionally has Metastases from Other
been considered the expression of systemic Primary Tumors
disease that requires chemotherapy; however,
liver-only metastatic disease occurs in 5% to Radiofrequency ablation has been performed
12% of breast cancer patients and they in several patients with cancer histologies dif-
may be eligible for surgical metastasectomy. ferent from colorectal and breast carcinoma. In
Early reports demostrated improved survival our experience RFA represents a useful treat-
(18-24% at 5 years' follow-up) in patients ment in patients with metastases from previ-
undergoing resection of limited hepatic metas- ously treated malignancies provided that local
tases from breast cancer (21-23). More recent control of liver disease may be beneficial from
reports yielded even superior survival rates the oncologic perspective in terms of improved
with 51 % with disease-free interval with stage survival or quality of life, and that size, number,
of primary as prognostic factor (24). and location allow adequately wide necrosis. In
Therefore, surgery may allow long-lasting our institution patients with liver metastases
benefit and be potentially curative; patients from neuroendocrine, gastric, pancreatic, renal,
with hepatic resection as the first and sole site pulmonary, uterine, and ovarian cancer and
of relapse have a threefold greater median sur- from melanoma have been treated successfully
vival than patients treated with standard non- with RFA.
surgical treatment. Under these conditions RFA has to be con-
With this background, we used percutaneous sidered a less invasive therapeutic alternative
RFA to achieve local control of liver metastases to surgery. Indications for RFA of liver metas-
in patients with breast cancer (30). Twenty-four tases from other primary cancers may follow
patents were treated for 64 liver metastases: those of surgical resection. Hepatectomy for
complete necrosis was obtained with a single liver metastases appears favorable in patients
treatment session in 92% of lesions. Interest- with gynecologic, gastric, and testicular primary
ingly, the rate of complete necrosis was superior tumors, with survival rates over 20%; unfortu-
to that obtained with colorectal metastases of nately, no definite selection criteria have been
comparable size (96% for lesions less than 3 cm reported in the literature (22,26,27).
and 75% for 3- to 5-cm lesions). These figures Particular consideration is reserved for
suggest that occult invasion of surrounding patients with neuroendocrine tumors, which
liver tissue may be absent or less pronounced commonly metastasize to the liver. In cases with
in breast tumors (30). metastatic disease, combinations of surgical
Over half of the patients (58%) developed debulking and medical treatment usually are
new metastases during follow-up, but 63% of 16 used. In patients with limited metastases,
patients with liver-only metastatic breast cancer surgery is considered a treatment option. Liver
24. Ablation of Liver Metastases 319

metastasectomy from endocrine primary 10. Semelka RC, HeImberger TC Contrast agents
tumors is followed by long survival in a sub- for MR imaging of the liver. Radiology 2001;
stantial proportion of patients with 56% to 74% 218:27-38.
5-year survival rates (22,25-27). Interesting II. Geoghegan JG, Scheele 1. Treatment of colorec-
results have been reported with RFA, used tal liver metastases. Br J Surg 1999;86:158-169.
12. Yoon SS, Tanabe KK. Surgical treatment and
alone or in combination with surgery, for local
other regional treatments for colorectal cancer
control of metastases from neuroendocrine liver metastases. Oncologist 1999;4:197-208.
tumors: disease eradication was obtained in 13. Taylor M, Forster J, Langer B, et al. A study
28.5% of patients treated with curative intent of prognostic factors for hepatic resection for
(29). colorectal metastases. Am J Surg 1997;173:467-
471.
References 14. Nordlinger B, Guigner M, Vaillant JC, et al. Sur-
gical resection of colorectal carcinoma metas-
1. Bhattacharya R, Rao S, Kowdley KV. Liver tases to the liver. A prognostic scoring system to
involvement in patients with solid tumors of improve case selection based on 1568 patients,
nonhepatic origin. Clin Liver Dis 2002;6:1033- Association Francaise de Chirurgie. Cancer 1996;
1043. 77:1254-1262.
2. Nordlinger B, Rougier P. Nonsurgical methods 15. Elias D, Cavalcanti A, Sabourin JC, et al. Resec-
for liver metastases including cryotherapy, radio- tion of liver metastases from colorectal cancer:
frequency ablation and infusional treatment: the real impact of the surgical margin. Eur J Surg
what's new in 200l? Curr Opin Oncol 2002;14: OncoI1998;24:174-179.
420-423. 16. Cady B, Jenkins RL, Steelde GD, et al. Surgical
3. Sica GT, Ji H, Ros PRo CT and MR imaging of margin in hepatic resection for colorectal metas-
hepatic metastases. AJR 2000;174:691-698. tasis: a critical and improvable determinant of
4. Valls C, Andia E, Sanchez A, et al. Hepatic outcome. Am Surg 1998;227:566-571.
metastases from colorectal cancer: preoperative 17. Holm A, Bradley E, Aldrete JS. Hepatic resec-
detection and assessment of resectability with tion of metastases from colorectal carcinoma.
helical CT. Radiology 200l;218:55-60. Morbidity, mortality and patterns of recurrence.
5. Harvey CJ, Blomley MJK, Eckersley RJ, et al. Ann Surg 1989;209:428-434.
Hepatic malignancies: improved detection with 18. Fong Y, Salo 1. Surgical therapy of hepatic
pulse-inversion US in the late phase of enhance- colorectal metastasis. Semin OncoI1999;26:514-
ment with SHU 508A-early experience. Radi- 523.
ology 2000;216:903-908. 19. Liu LX, Zhang WH, Jiang HC Current treat-
6. Albrecht T, Hoffmann CW, Schmitz SA, et al. ment for liver metastases from colorectal cancer.
Phase-inversion sonography during the liver World J GastroenteroI2003;9:193-200.
specific late phase of contrast enhancement: 20. Hugh TJ, Kinsella AR, Poston G1. Management
improved detection of liver metastases. AJR strategies for colorectalliver metastases-part I.
2001;176:1191-1198. Surg OncoI1997;6:19-30.
7. Quaia E, Bertolotto M, Forgacs B, et al. Detec- 21. Elias D, Lasser PH, Montrucolli D, Bonvallo S,
tion of liver metastases by pulse inversion Spielmann M. Hepatectomy for liver metastases
harmonic imaging during Levovist late phase: from breast cancer. Eur J Surg Oncol 1995;21:
comparison with conventional ultrasound and 51D-513.
helical CT in 160 patients. Eur Radiol 2003;13: 22. Elias D, Cavalcanti A, Eggenspieler P, et al.
475-483. Resection of liver metastases from a noncolo-
8. Solbiati L, Tonolini M, Cova L, et al. The role of rectal primary: indications and results based on
contrast-enhanced ultrasound in the detection 147 monocentric patients. J Am Coll Surg 1998;
of focal liver lesions. Eur Radiol 200l;11:E15- 187:487-493.
E26. 23. Raab R, Nussbaum KT, Behrend M, et al. Liver
9. Albrecht T, Blomley MJ, Burns PN, et al. metastases of breast cancer: results of liver resec-
Improved detection of hepatic metastases with tion. Anticancer Res 1998;18:2231-2234.
pulse-inversion US during the liver-specific 24. Maksan SM, Lehnert T, Bastert G, et al. Curative
phase of SHU 508A: multicenter study. Radiol- liver resection for metastatic breast cancer. Eur
ogy 2003;227:361-370. J Surg Oncol 2000;26:209-212.
320 L. Solbiati et al

25. Chen H, Hardacre JM, Uzar A, et al. Isolated primary and secondary liver tumors. Am J Surg
liver metastases from neuroendocrine tumors: 1999;177:303-306.
does resection prolong survival? J Am Coll Surg 39. Lencioni R, Goletti 0, Arrnillotta N, et al.
1998;187:88-93. Radiofrequency thermal ablation of liver metas-
26. Berney T, Mentha G, Roth AD, et al. Results of tasis with cooled-tip electrode needle: results of
surgical resection of liver metastases from non- a pilot clinical trial. Eur Radiol 1998;8:1205-
colorectal primaries. Br J Surg 1998;85:1423- 1211.
1427. 40. Rose DM, Allegra DP, Bostick PJ, et al.
27. Lindell G, Ohlsson B, Saarela A, et al. Liver Radiofrequency ablation: a novel primary and
resection of noncolorectal secondaries. J Surg adjunctive ablative technique for hepatic malig-
Oncol1998;69:66-70. nancies. Am Surg 1999;65:1009-1014.
28. Maksan SM, Lehnert T, Bastert G, Herfarth C. 41. Rossi S, Buscarini E, Garbagnati F, et al. Percu-
Curative liver resection for metastatic breast taneous treatment of small hepatic tumors by
cancer. Eur J Oncol 2000;26:209-212. an expandable RF needle electrode. AJR 1998;
29. Hellman P, Ladjevardi S, Skogseid B, et al. 170:1015-1022.
Radiofrequency tissue ablation using cool tip for 42. De Baere T, Elias D, Dromain C, et al. Radiofre-
liver metastases of neuroendocrine tumors. quency ablation of 100 hepatic metastases with
World J Surg 2002;26:1052-1056. a mean follow-up of more than 1 year. AJR 2000;
30. Livraghi T, Goldberg SN, Solbiati L, et al. Percu- 175(6):1619-1625.
taneous radiofrequency ablation of liver metas- 43. Goldberg SN, Gazelle GS, Solbiati L, et al.
tases from breast cancer: initial experience in 24 Radiofrequency tissue ablation: increased lesion
patients. Radiology 2001;220:145-149. diameter with a perfusion electrode. Acad
31. Goldberg SN, Gazelle GS, Mueller PRo Thermal Radiol 1996;3:636-644.
ablation therapy for focal malignancy. A unified 44. Goldberg SN, Solbiati L, Hahn PF, et al. Large-
approach to underlying principles, techniques, volume tissue ablation with radiofrequency by
and diagnostic imaging guidance. AJR 2000;174: using a clustered, internally cooled electrode
323-331. technique: laboratory and clinical experience
32. Solbiati L, Goldberg SN, Ierace T, et al. Hepatic in liver metastases. Radiology 1998;209:371-379.
metastases: percutaneous radio-frequency abla- 45. Goldberg SN, Stein MC, Gazelle GS, et al. Per-
tion with cooled-tip electrodes. Radiology 1997; cutaneous radiofrequency tissue ablation: opti-
205:367-373. mization of pulsed-radiofrequency technique
33. Solbiati L, Ierace T, Goldberg SN, et al. Percuta- to increase coagulation necrosis. J Vase Interv
neous US-guided radiofrequency tissue ablation Radiol1999;10:907-916.
of liver metastases: treatment and follow-up in 46. Livraghi T, Goldberg SN, Monti F, et al. Saline-
16 patients. Radiology 1997;202:195-203. enhanced radiofrequency tissue ablation in the
34. Livraghi T, Goldberg SN, Lazzaroni S, et al. treatment of liver metastases. Radiology 1997;
Small hepatocellular carcinoma: treatment with 202:205-210.
radiofrequency ablation versus ethanol injec- 47. De Baere T, Bessoud B, Dromain C, et al. Per-
tion. Radiology 1999;210:655--661. cutaneous radiofrequency ablation of hepatic
35. Livraghi T, Goldberg SN, Lazzaroni S, et al. tumors during temporary venous occlusion. AJR
Hepatocellular carcinoma: radio-frequency abla- 2002;178:53-59.
tion of medium and large lesions. Radiology 2000; 48. Rhim H, Goldberg SN, Dodd GD III, et al.
214:761-768. Essential techniques for successful radio-
36. Solbiati L, Livraghi T, Goldberg SN, et al. Per- frequency thermal ablation of malignant hepatic
cutaneous radiofrequency ablation of hepatic tumors. Radiographies 2001;21 (spec. no.):S17-35.
metastases from colorectal cancer: long-term 49. Goldberg SN, Kamel IR, Kruskal JB, et al. Radio-
results in 117 patients. Radiology 2001;221: frequency ablation of hepatic tumors: increased
159-166. tumor destruction with adjuvant liposomal
37. Curley SA, Izzo F, Delrio P, et al. Radiofre- doxorubicin therapy. AJR 2002;179:93-101.
quency ablation of unresectable primary and 50. Akhurst T, Larson SM. Positron emission tomog-
metastatic hepatic malignancies: results in 123 raphy imaging of colorectal cancer. Semin Oncol
patients. Ann Surg 1999;230:1-8. 1999:26(5):577-583.
38. Jiao LR, Hansen PD, Havlik R, et al. Clinical 51. Anderson GS, Brinkmann F, Soulen MC, et al.
short-term results of radiofrequency ablation in FDG positron emission tomography in the sur-
24. Ablation of Liver Metastases 321

veillance of hepatic tumors treated with radio- 53. Livraghi T, Solbiati L, Meloni F, et al. Percuta-
frequency ablation. Clin Nucl Med 2003;28: neous radiofrequency ablation of liver metas-
192-197. tases in potential candidates for resection: the
52. Elias D, De Baere T, Smayra T, et al. Percuta- "test of time" approach. Cancer 2003;97:3027-
neous radiofrequency thermoablation as an 3035.
alternative to surgery for treatment of liver
tumour recurrence after hepatectomy. Br J Surg
2002;89:752-756.
25
Ablation for Hepatocellular Carcinoma
Maria Franca Meloni and Tito Livraghi

Hepatocellular carcinoma (HCC) is the fifth The range of treatment options is fairly wide,
most common cancer in the world, and it rep- and the choice is not always easy, given the
resents more than 5% of all cancers (1). It number of variables to be assessed. The options
usually coexists with underlying chronic today are surgery (liver transplantation and
liver disease, and according to the stage, one partial resection); percutaneous ablation tech-
disease will prevail over the other. The stage niques (ethanol injection, thermal ablation);
of HCC at the time of diagnosis is very intraarterial techniques including transarterial
important to decide the type of treatment- embolization and transarterial chemoemboliza-
possibly curative when the diagnosis is early, tion (TAE-TACE); radiation; and systemic
palliative or no treatment when the diagnosis is therapies. Most of them have not been vali-
late. dated with randomized controlled studies, that
In countries in which ultrasound (US) is, versus the natural history or versus other
screening of the population at risk is practiced, therapies, but are applied only on the basis of
early diagnosis of HCC is possible. Elsewhere, local control of the tumor.
the diagnosis is not made until the disease is The only therapy likely to afford a definitive
already advanced. Because HCC is an organ cure in selected patients is liver transplantation
pathology, the first nodule detected is only a (LT). However, the shortage of donors, high
prelude to others; thus HCC needs multistep costs, advanced technology, and ethical reasons
detection and treatment. limit this option. Surgical resection can achieve
The survival studies of untreated patients complete ablation of the first nodule (also of
with HCC that usually are taken as points peripheral satellites if anatomic resection is
of reference fall into four conventional and carried out). According to the Japanese Nation-
gross groups: (1) the disease is at an early wide Survey in 2001, re-resection is performed
stage (single HCC <Scm, or up to three for only 1.6% of new lesions (3).
lesions <3 cm) and therefore likely to be In the majority of studies, the survival rates
amenable to cure; (2) the disease is considered after surgery were roughly comparable with
intermediate, that is, without an invasive tumor those obtained with percutaneous ethanol
pattern (absence of portal thrombosis or extra- injection therapy (PElT) (4,5). A more recent
hepatic spread), and asymptomatic; (3) the trial also documented similar results (6). In this
disease is advanced (between intermediate and latter study, 97 comparable patients with HCC
terminal stage); and (4) terminal (PST stage 3 <3 cm in diameter and three or fewer in number
or 4 or Okuda stage III). A similar staging has were treated, 39 with PEl and 58 with surgical
been used for practical purposes in many resection, respectively, during the same period.
centers, but was only recently codified by the Their 3- and 5-year survival was 82% and 59%
Liver Unit of Barcelona (2). in the PElT group, and 84 % and 61 % in the

322
25. Ablation for Hepatocellular Carcinoma 323

surgery group. These results probably reflect a satellites after complete necrosis of the domi-
balance between the advantages and disadvan- nant lesion.
tages of the two therapies. Moreover, the results
of surgery have been hampered by an incorrect
selection of patients, with some undergoing Rationale for Percutaneous
resection, even though they have adverse prog- Interventional Procedures in
nostic factors. PElT survival curves are always
better than those of resected patients who
HCC
present with adverse prognostic factors.
The following are reasons for use of percuta-
Another confirmation of such an interpreta-
neous interventional procedures for tumor
tion comes from the report of the Liver Cancer
ablation:
Study Group of Japan. Three- and 5-year sur-
vival in Child's class A patients with a single 1. They do not have the disadvantage of
HCC <5 cm was 67% and 49% respectively, in loss of or damage to nonneoplastic hepatic
the previous report of 1982-89 when PElT was parenchyma. The underlying chronic hepatic
not available. Survival was 77% and 59% in disease, generally of viral origin, accompanies
the report of 1988-95 (75% and 46% with the neoplastic disease in its different stages.
PElT) because of better patient selection (peri- Percutaneous therapies should not worsen liver
operative mortality remained substantially function.
unchanged) and the probable shift to PElT of 2. They are low-risk procedures. In pub-
some patients who, before the advent of PElT, lished series, the mortality rates are extremely
would have been treated surgically (7). low: 0.09% in the largest study of PElT (12),
After the introduction of other percutaneous ranging between 0.3% and 0.5% in the largest
techniques, mainly radiofrequency ablation studies of RFA (11,13), and no mortality due to
(RFA), a question that remains is the choice selected TAE (14,15).
between PElT and RFA. We consider these 3. They can be easily repeated when new
techniques complementary as well as selective lesions appear, as occurs in most patients within
TACE. We use them according to the features 5 years. Hepatocellular carcinoma is considered
of the disease, most notably number, location of an organ disease, since it has been proven to be
tumor(s), presence of satellites, portal throm- multicentric over time. A recent study demon-
bosis, and response to prior treatment. This strated that multicentricity is already present in
approach means a tailored strategy for every 50% of early stages, and that 93% of resected
patient, that is, a multistep multimodality patients with a single small HCC developed
image-guided local treatment. For instance, other lesions within 5 years (16). Since new
multifocal HCC can be treated with only lesions reflect the natural history of the disease,
one, all, or a combination of the techniques. the patient should be followed frequently so
Most lesions are treated with RFA, since RFA that the new lesions can be treated as they are
obtains both a higher rate of necrosis than detected. An advantage is that the patient can
PElT and a higher rate of necrosis and be followed by the same physician in the diag-
fewer side effects than TACE (8,9). But PElT nostic as well as therapeutic phase.
is preferable in tumors at risk for RFA, such 4. The low cost and easy availability of the
as those adjacent to main biliary ducts necessary material (particularly as regards
because of the possibility of stricture, and PElT) and the simplicity of these techniques
those juxtaposed to gastrointestinal structures make it possible to perform them even in
because of the possibility of perforation (10,11), peripheral centers. Patients with small HCCs
or for treating segmental portal thrombosis. generally are treated as outpatients, and most
Selective TACE is used for lesions not patients quickly return to normal daily life. In
recognizable at ultrasound, in lesions not com- Italy, the cost of one PElT cycle is about $1000,
pletely necrosed and not re-treatable by RFA partial resection about $30,000, and liver trans-
or PElT, and in the presence of peripheral plantation about $125,000 (17). In Japan, an
324 M.E Meloni and T. Livraghi

average cost is $759 for outpatient PElT, and Other Injectable Therapies
$27,105 for resection (18). Since every year the
number of patients who develop the disease is Honda et al (23) treated 20 patients with 23
about 25,000 in Japan and 10,000 in Italy, the lesions less than 3 cm in diameter with hot
problem of cost is an essential consideration saline solution therapy. No complications
(17). The cost of RF generators ranges from occurred. Therapeutic efficacy was evaluated
$12,000 to $30,000. Nonreusable needle elec- with CT, histopathologic examination, and
trodes cost $500 to $1000. The cost of TACE is serum marker levels.
about $1000 per session (19). Ohnishi et al (24) treated 91 patients with
small HCCs using percutaneous injection of
acetic acid (from 15% to 50% concentration)
under US guidance. No major complications
Percutaneous Interventional were reported. No evidence of local recurrence
Procedures was found 6.5 years later. Survival at 1,2,3,4,
and 5 years was 95%, 87%, 80%, 63%, and
Percutaneous interventional techniques can be 49%, respectively. The disease-free survival at
performed using an approach directly through 1,2,3,4, and 5 years was 83%, 54%, 50%, 37%,
the liver parenchyma or through the hepatic and 29%. A second study reported from the
artery. Interstitial local ablation therapies are same group, using percutaneously injected 50%
those treatment modalities that allow the acetic acid in 25 patients with HCC, demon-
introduction of a damaging agent directly into strated shrinkage of all lesions (25). No viable
the neoplastic tissue. Local ablation therapies tumor was found at follow-up by biopsy, CT, or
may be based on destroying the tissue surgical wedge resection (two cases).
chemically, such as with ethyl alcohol (PElT) or A comparison among PElT, acetic acid, and
acetic acid, or physically, as with laser, radio- hot saline injection has not been reported.
frequency (RF), or microwave. The range of
indications for local ablation treatments is
becoming wider than for surgery or intraarter- Radiofrequency Ablation (RFA)
ial therapies. Thermoablation with RFA exploits the conver-
sion of energy of an electromagnetic wave into
heat. The most widely used instruments are
Percutaneous Ethanol Injection
made by three companies: Radiotherapeutics,
Local-regional therapies may be based on Sunnyvale, CA; RITA Medical Systems, Moun-
destroying the tissue chemically, such as with tain View, CA; and Radionics, Burlington, MA
ethyl alcohol (PElT). It was the first percuta- (26,27). Each of the devices uses a different
neous treatment to be used (20). For some needle design, wattage, and algorithms. The first
years, only patients with up to three small two use an expandable electrode that is 1.9mm
«5 cm) lesions were treated, and this guideline in diameter, which, once positioned in the
still applies at many centers. With the introduc- tumor, opens out into seven to 10 retractable
tion of the "single-session" procedures under curved electrodes around the target, like an
general anesthesia (21), even patients with umbrella. These systems produce a repro-
lesions greater in number or larger in size are ducible area of necrosis about 3 to 4cm in diam-
now treated. Percutaneous ethanol injection eter. To increase the necrotic area with the
therapy spreads rapidly, thanks to its ease of aforementioned techniques, interruption of the
execution, safety, low cost, repeatability, and arterial supply to the tumor by means of occlu-
therapeutic efficacy. Survival rates are compa- sion of the hepatic artery with a balloon
rable to those of surgical resection (4,18,22). catheter or the feeding arteries with gelatin
Today PElT is one of the treatments most fre- sponge particles can be performed (28).
quently offered to patients detected by US The third device utilizes a cold perfusion elec-
screening to have HCC. trode with a diameter of 1.2 mm, and an exposed
25. Ablation for Hepatocellular Carcinoma 325

tip of 2 to 3 cm (29). By avoiding early incre- Benefiting from the "oven effect," a fourth
ments of impedance linked to carbonization, study, on the treatment of larger HCCs, was
these electrodes permit application of a greater performed (33). In 114 patients with 80
power than conventional electrodes. To obtain medium-diameter (3.0-5.0cm) or 40 large
cooling, a physiologic solution brought to 2° to HCCs (5.1-9.0cm diameter), complete necrosis
SoC is circulated within two coaxial lumens that was attained in 60 lesions (47.6%) and nearly
are situated in the electrode. The technique pro- complete necrosis (90-99%) in 40 other lesions
duces a reproducible area of necrosis of 2.4 cm (31.7%). Medium and non-infiltrating tumors
in diameter. A recently constructed electrode were treated successfully (71 %) more often
with three cooled tips that permits a higher than large and infiltrating tumors (23%). Two
current deposition yields greater than 4.5 cm major complications (death from septic shock
diameter of coagulation necrosis (30). due to peritonitis in an obese patient with dia-
Unexpectedly, it has been observed that in betes and hemorrhage that required laparot-
HCCs of about 3 cm in diameter, the area and omy) and five minor complications were
shape of the necrosis reproduced that of the observed (11). The results of the study sup-
original tumor like a mold. Such an effect was ported the importance of the "oven effect".
designated the "oven effect". It was believed However, the oven effect also may explain the
that the surrounding cirrhotic tissue with its limited success in treating satellite nodules that
high fibrotic component and poor vasculariza- remain a limitation for RFA or PElT ablation,
tion was a poor conductor and thus functioned despite continued technical improvements.
as an insulating material. These factors resulted Probably, the fibrotic tissue that is interposed
in higher deposition of energy within the neo- between the main tumor and the satellites
plastic tissue (8). limits heat diffusion. A longer follow-up is
Studies have been published on the treatment needed to determine the prognostic improve-
of HCC with RFA on local therapeutic efficacy. ment that results from treatment of the main
The first was carried out using a hooked tumor alone. However, in some cases, when the
expandable electrode on 23 patients with HCC location of satellites is favorable, RFA can
up to 3.5 cm in diameter. With an average of 1.4 obtain satisfying results.
sessions, a complete response was reported in
all tumors and no complications were observed
(31). The second study was controlled and
Microwave
prospective; it compared PElT and RFA on 86 Microwave therapy was devised by a Japanese
patients with 112 HCCs that measured up to team in 1986 (34). It is the next thermal therapy
3cm in diameter. A complete response was used for interstitial percutaneous ablation.
achieved in 90.3% with RFA and in 80.0% with The equipment consists of a microwave gener-
PElT. These results were obtained with an ator and an 18-gauge reusable needle electrode
average of 1.2 sessions for RFA and 4.8 sessions that is 25 cm long. The electrode is placed
for PElT. However, there were more complica- through a 14-gauge styleted access needle. The
tions with RFA, that is, one severe (hemothorax microwave needle functions as an antenna for
that required drainage) and four minor, com- externally applied energy at 1000 to 2450MHz
pared to none with PElT (8). The third study (35).
was prospective, and it compared PElT and Saitsu et al (36) published the first report on
RFA on 119 patients with a single HCC that intraoperative microwave coagulation treat-
measured up to 3 cm in diameter. Complete ment in 21 patients with HCCs less than 5 cm
tumor necrosis was achieved in 100% of the in diameter. Recurrence of disease occurred in
RFA group, and in 94 % of the PElT group, with 23.8% of the lesions. Twenty patients survived
the same local recurrence rate. But RFA to a maximum of 39 months. Several studies
required an average of only 1.5 sessions, with good clinical results from treatment of
whereas PElT required an average of 4.0 ses- liver tumors with microwave have been pub-
sions (32). lished (37-41). The results of 60 patients with
326 M.P. Meloni and T. Livraghi

69 small HCCs treated by microwave and Cryoablation


studied by dynamic CT demonstrated a com-
plete response in 72 %, and incomplete necro- Cryoablation is the oldest interstitial ther-
sis or tumor recurrence in 28 %. The mean motherapy to treat liver tumors (51). It uses
disease-free period was 24.2 months. The sur- intraoperative cooled probes that measure
vival rates at 1 and 2 years were 83.1 % and 3 mm in diameter to freeze and destroy large
68.7%, respectively. Major complications were areas of tumor tissue (52). This technique has
ascites in three patients, pleural effusion in two, recently became adapted for percutaneous use
intraperitoneal abscess in one, and seeding in (53).
two (38,39).
Selective Transarterial
Interstitial Laser Photocoagulation Chemoembolization
Interstitial laser photocoagulation is another For many years, TAE or TACE of the whole
thermotherapy that induces coagulation necro- liver was the most widely used therapeutic
sis in tumor cells (42). The first studies on liver option for patients with intermediate HCC. The
tumors (hepatoma and metastases) with laser rationale for this approach is that in contrast
therapy were reported by Hashimoto et al (43) with normal liver tissue, HCCs receive almost
and Steger et al (44). the totality of their blood supply through
Laser therapy is performed with US- or CT- the hepatic artery. Transarterial embolization
guided imaging, using conscious sedation and and TACE have marked antitumoral effect,
local lidocaine anesthesia. Laser fibers are although only in encapsulated lesions. How-
inserted through a cannula and attached to a ever, recent randomized controlled trials did
ne odymi um :yttri um- al umin um- garnet not show any statistically significant difference
(Nd:YAG) laser, which is excited at 2 to 4 W for in survival between treated and untreated
5 to 15 minutes. patients (54-56). Treated patients have slower
Several reports have been published on the tumor progression and even a decrease in the
use of laser coagulation to treat liver tumors incidence of portal vein invasion; however,
(45-50). Lees and Gillams's group (19) re- cancer-related complications or liver decom-
ported on patients with inoperable colorectal pensation was no different compared to
metastases to the liver. They had a 5-year untreated patients. This is probably due to a
survival rate of 26%, and a median survival of counterbalance between local tumor control
27 months; these results were comparable to and damage to nonneoplastic tissue, which
patients who had been operated and had a hastens liver insufficiency, even though a study
median survival of 33 months and a 5-year sur- particularly dedicated to this problem failed to
vival rate of 30%. In 500 patients, major com- demonstrate such an interconnection (57).
plications have included segmental infarction In a study by the Liver Unit of Barcelona, 2-
in five patients, abscesses in three patients, and 4-year survival was 49% and 13% after
pleural effusion in one patient, tumor seeding TAE, and 50% and 27% after no treatment
in six patients, pain, and one death from a (56). A more recent randomized controlled trial
spontaneous hepatic infarction that occurred 6 (58) demonstrated improved survival in strin-
days after treatment. A recent study in 74 gently selected patients with intermediate-
patients with 92 small HCCs demonstrated stage HCC: 3-year survival was 29% in the
complete ablation in 97% of lesions, with local treated group and 0% in the untreated group.
recurrence rates (1-5 years) from 1.6% to Thus, the current trend is to use TAE or TACE
6%. One-, 3-, and 5-year survival rates were with lobar disease by selective catherization
99%, 68%, and 15%, respectively. No major because it does not cause deterioration of liver
complications occurred (50). Interstitial laser function, compared to the whole-liver tech-
photocoagulation is a safe therapy for small nique, and with a better tumor response being
HCCs. achieved (59). A recent meta-analysis study
25. Ablation for Hepatocellular Carcinoma 327

demonstrated that chemoembolization im- plastic tissue, or when the same picture is
proves survival of patients with unresectable confirmed with ultrasound contrast media
HCC, and may become a standard treatment examination, and on scans performed on suc-
(60). cessive follow-up visits. The absence of contrast
media enhancement means the absence of
blood flow due to necrosis and fibrotic effects.
Evaluation of Therapeutic Even with such characteristics, after the treat-
Efficacy ment, necrotic tumor occupies space and
remains visible in place of the tumor, but
To evaluate therapeutic response means to reduces in size over time. Ultrasound with con-
determine whether a tumor has become com- trast agents can be useful, but should not be
pletely necrotic or whether areas of neoplastic used as the only test to establish the result,
tissue remain. Measures to assess response because it is less sensitive than CT in detecting
include a combination of investigations and vascularity of small residual tumor areas (61).
serum assays for tumor markers. These metrics Different degrees of Lipiodol uptake after
are the same as those used during initial TAE-TACE are a good indicator of therapeu-
staging. We prefer to use spiral CT with tripha- tic efficacy, and its labeling can be checked
sic technique (4-5 mL/sec; 20, 60, and 300 during follow-up by CT scans (64). Re tumor
seconds after the injection of contrast medium), markers, we use a-fetoprotein (AFP) and des-
and US with first- and second-generation ultra- gamma-carboxy-prothrombin (DCP), which
sound contrast agents (USCAs) (Levovist, often are complementary. Nevertheless, their
Shering, Berlin, Germany; SonoVue, Bracco, assay is useful only if levels initially are high.
Milan, Italy). A recent study demonstrated that When imaging techniques show a complete
the sensitivity of pulse inversion harmonic response that is not corroborated by reduction
ultrasound imaging (83.3%) with Levovist was in AFP or DCP levels, it means that neoplastic
significantly greater (p <.05) for detecting re- tissue is not detected or is growing elsewhere
sidual nonablated tumor after radiofrequency outside the liver. Moreover, an increase in
treatment compared to conventional contrast- levels during follow-up always suggests a local
enhanced power Doppler sonography (61). recurrence or the appearance of new lesions.
Solbiati et al (62) reported that the routine use Follow-up with ultrasound alone, or with con-
of contrast-enhanced harmonic US (SonoVue) trast agents, with CT, and serum assay of tumor
reduces costs by decreasing the number markers is done a month after treatment and
of percutaneous ablations and follow-up then every 4 to 6 months.
examinations.
Other imaging examinations (MRI, PET) or
biopsy are performed only in cases of doubt Multistep Modality
about partial or complete response. If the areas
of viable tissue are very small, beyond the Image-Guided Local Treatment:
present powers of imaging resolution, they will Why and When
obviously not be recognizable. Some authors
find it difficult to recognize viable tissue from The large number of patients enrolled in US
coagulation necrosis when the target is small, screening programs has created a demand
and consequently there is uncertainty about re- for effective, safe, repeatable, and economical
treatment (63). However, residual tumor will be treatment that can be made available in many
more easily identified during follow-up imaging centers. In the absence of randomized trials, it
examinations as zones of increased enhance- is and will be difficult to find agreement on indi-
ment at CT or of increased growth of enhanced cations of the respective therapeutic options.
tumor. The response is considered complete on In our opinion, the only course at this time is
imaging when CT scans show total disappear- to extrapolate from retrospective comparative
ance of contrast enhancement within the neo- studies and from reports on prognostic factors
328 M.E Meloni and T. Livraghi

the unequivocal information that can prevent and a tumor less than 2cm in diameter, when
useless or even damaging therapies. Moreover, the possibility of peritumoral microinvasion is
economic resources and the expertise that is low, and the comparative rate of complete abla-
available at each center are other factors that tion with percutaneous ablation techniques is
have a role in the choice of treatment. probably 100%.
In our opinion, multistep modality image The best strategy to follow might be early
guidance of local treatment is indicated as the detection of HCCs by means of US surveillance
treatment of choice for most patients who in an at-risk population, treatment with per-
are followed by ultrasound screening, excluding cutaneous interventional techniques, follow-up
those candidates for partial resection, and who with imaging methods and tumor markers, and
have (1) favorable factors: Child's A class, (2) further treatment of any new lesions.
transaminases <3x normal values, (3) younger
than 75 years, (4) no portal hypertension, (5) References
normal bilirubin, (6) no portal thrombosis, (7)
single lesion (also with peripheral satellites), 1. Parkin DM, Bray F, Ferlay J, Pisani P. Estimating
the world cancer burden: GLOBOCAN 2000. Int
(8) feasibility of anatomic operation, and (9)
J Cancer 2001;94:153-156.
skilled operators assuring perioperative mor- 2. Llovet JM, Bru C, Bruix 1. Prognosis of hepato-
tality of less than 2%. However, we prefer to cellular carcinoma: the BCLC staging classifica-
perform RFA in single HCCs <2cm, since the tion. Semin Liver Dis 1999;19:329-339.
possibility of peritumoral microinvasion is low, 3. Arii S, Teramoto K, Kawamura T, et al. Charac-
and the rate of complete ablation approaches teristics of recurrent hepatocellular carcinoma in
100%. Furthermore, in these cases, the man- Japan and our surgical experience. J Hepatobil-
agement of patients following the rationale of iary Pancreat Surg 2001;8:397-403.
the "test of time" (based on survival benefit 4. Livraghi T, Bolondi L, Buscarini L, et al. No
from hepatic resection) is determined by the treatment, resection and ethanol injection in
biologic features of the tumor, rather than by hepatocellular carcinoma: a retrospective analy-
sis of survival in 391 patients with cirrhosis.
early detection. Resection could be performed J Hepatol 1995;22:522-526.
in patients whose tumors are incompletely 5. Ryu M, Shimamura Y, Kinoshita T, et al. Jpn J
treated with the multimodality approach, and Clin OncoI1997;27:251-257.
who do not develop additional tumors during 6. Yamamoto J, Okada S, Shimada K, et al. Treat-
follow-up. ment strategy for small hepatocellular carci-
noma: comparison of long-term results after
percutaneous ethanol injection therapy and sur-
Conclusions gical resection. Hepatology 2001;34:707-713.
7. The Liver Cancer Study Group of Japan. Pre-
The tailored combination of RFA, PElT, and dictive factors for long-term prognosis after
selective TACE is indicated as the treatment of partial hepatectomy for patients with hepatocel-
choice for most patients enrolled by US screen- lular carcinoma in Japan. Cancer 1994;74:2772-
ing, excluding those candidates for liver trans- 2780.
plantation and for partial surgical resection. 8. Livraghi T, Goldberg N, Lazzaroni S, Meloni F,
Unfortunately, transplantation is available for Solbiati L, Gazelle S. Small hepatocellular carci-
few patients in Western countries because of noma: treatment with radio-frequency ablation
versus ethanol injection. Radiology 1999;210:
the shortage of donors, in poorer countries
655-661.
because of the lack of advanced technology, and 9. Livraghi T, Meloni F, Corso R, Rampoldi A.
in Japan because of ethical reasons. Candidates Comparison between RF ablation and TACE in
for partial resection should not have adverse the treatment of multifocal hepatocellular carci-
prognostic factors (7,65,66). noma: preliminary experience. Radiology 2000;
It is still controversial whether a complex 217:287-288.
resection is an appropriate choice for a patient 10. Meloni MF, Goldberg SN, Moser V, Piazza G,
who does not have adverse prognostic factors Livraghi T. Colonic perforation and abscess fol-
25. Ablation for Hepatocellular Carcinoma 329

lowing radiofrequency ablation treatment of 23. Honda N, Guo Q, Uchida H, Ohishi H, Hiasa Y.
hepatoma. Eur J Ultrasound 2002;15(1-2):73-76. Percutaneous hot saline injection therapy for
11. Livraghi T, Solbiati L, Meloni F, Gazelle GS, hepatic tumors: an alternative to percutaneous
Halpern EF, Goldberg SN. Percutaneous radio- ethanol injection therapy. Radiology 1994;190(1):
frequency ablation: complications encountered 53-57.
in a multicenter study of the treatment of focal 24. Ohnishi K, Ohyama N, Ito S, Fujiwara K. Small
liver tumors. Radiology 2003;226:441--451. hepatocellular carcinoma: treatment with US-
12. Di Stasi M, Buscarini L, Livraghi T, et al. Percu- guided intratumoral injection of acetic acid.
taneous ethanol injection in the treatment of Radiology 1994;193(3):747-752.
hepatocellular carcinoma: a multicenter survey 25. Ohnishi K, Normura F, Ito S, Fujiwara K. Prog-
of evaluation practice and complication rates. nosis of small hepatocellular carcinoma (less
Scand J GastroenteroI1997;32:1168-1173. than 3cm) after percutaneous acetic acid injec-
13. Mulier S, Mulier P, Ni Y, et al. Complications of tion: study of 91 cases. Hepatology 1996;23(5):
radiofrequency coagulation of liver tumors. Br J 994-1002.
Surg 2002;89:1206-1222. 26. Gazelle GS, Goldberg SN, Solbiati L, Livraghi T.
14. Matsui 0, Kadoja M, Yoshikawa J, Gabata T, Thmor ablation with radiofrequency energy.
Takashima T, Demachi H. Small hepatocellular Radiology 2000;217:633-646.
carcinoma: treatment with subsegmental arterial 27. McGahan JP, Dodd GD III. Radiofrequency
embolization. Radiology 1993;188;78-93. ablation of the liver: current status. AJR 2001;
15. Uchida H, Matsuo N, Nishimine K, Nishimine Y, 176:3-16.
Sakaguchi H, Ohishi H. Transcatheter arterial 28. Rossi S, Garbagnati F, Lencioni R, et al. Percu-
embolization for hepatoma with lipiodol. taneous radiofrequency ablation of nonresect-
Hepatic arterial and segmental use. Semin Inter- able hepatocellular carcinoma after occlusion of
vent RadioI1993:l0:19-26. tumor blood supply. Radiology 2000;217:119-
16. Nakashima 0, Kojiro M. Recurrence of hepato- 126.
cellular carcinoma: multicentric occurrence or 29. Goldberg SN, Gazelle GS, Solbiati L, Rittman
intrahepatic metastasis? A viewpoint in terms of WJ, Mueller PRo Radiofrequency tissue ablation:
pathology. J Hepatobiliary Pancreat Surg 2001; increased lesion diameter with a perfusion elec-
8:404--409. trode. Acta RadioI1996;3:636-644.
17. Livraghi T, Giorgio A, Marin G, et al. Hepato- 30. Goldberg SN, Solbiati L, Hahn PF, et al. Large
cellular carcinoma and cirrhosis in 746 patients: volume tissue ablation with radiofrequency by
long-term results of percutaneous ethanol injec- using a clustered, internally cooled electrode
tion. Radiology 1995;197:101-108. technique: laboratory and clinical experience in
18. Kotoh K, Sakai H, Sakamoto S, et al. The effect liver metastases. Radiology 1998;209:371-379.
of percutaneous ethanol injection therapy on 31. Rossi S, Buscarini E, Garbagnati F. Percutaneous
small solitary hepatocellular carcinoma is com- treatment of small hepatic tumors by an expand-
parable to that of hepatectomy. Am J Gastroen- able RF needle electrode. AJR 1998;170:1015-
teroI1994;89:194-198. 1022.
19. Dodd GD III, Soulen MC, Kane RA, et al. Min- 32. Ikeda M, Okada S, Ueno H, Okusaka T,
imally invasive treatment of malignant hepatic Kurijama H. Radiofrequency ablation and
tumors: the threshold of a major breakthrough. percutaneous ethanol injection with small
Radiographies 2000;20(1):9-27. hepatocellular carcinoma: a comparative study.
20. Livraghi T, Festi D, Monti F, Salmi A, Vettori C. Jpn J Clin OncoI2001;31:322-326.
US-guided percutaneous alcohol injection of 33. Livraghi T, Goldberg SN, Lazzaroni S, et al.
small hepatic and abdominal tumors. Radiology Hepatocellular carcinoma: radiofrequency abla-
1986;161:309-312. tion of medium and large lesions. Radiology
21. Livraghi T, Vettori C, Torzilli G, Lazzaroni S, 2000;214:761-768.
Pellicano S, Ravasi S. Percutaneous ethanol 34. Tabuse Y, Tabuse K, Mori K, et al. Percutaneous
injection of hepatic tumors: single session microwave tissue coagulation in liver biopsy:
therapy under general anesthesia.AJR 1993;160: experimental and clinical studies. Nippon Geka
1065-1069. Hokan 1986;55:381-392.
22. Onodera H, Ukai K, Nakano N, et al. Outcomes 35. King RWP, Shen LC, Wu TT. Embedded insu-
of 116 patients with hepatocellular carcinoma. lated antenna for communication and heating.
Cancer Chemother PharmacoI1994;33:103-108. Electromagnetics 1981;1:51-172.
330 M.P. Meloni and T. Livraghi

36. Saitsu H, Mada Y, Taniwaki S, et al. Investigation 48. Pacella CM, Bizzarri G, Ferrari FS, et al.
of microwave coagulo-necrotic therapy for 21 Interstitial photocoagulation with laser in the
patients with small hepatocellular carcinoma less treatment of liver metastases. Radiol Med 1996;
than 5 cm in diameter. Nippon Geka Gakkai 92:438-447.
Zasshi 1993;94(4):359-365. 49. Vogi TJ, Mack MG, Roggan A, et al. Internally
37. Seki T, Wakabayashi M, Nakagawa T, et al. cooled power laser for MR-guided interstitial
Ultrasonically guided percutaneous microwave laser-induced thermotherapy of liver lesions:
coagulation therapy for small hepatocellular initial clinical results. Radiology 1998;209:
carcinoma. Cancer 1994;74(3):817-825. 381-385.
38. Murakami R, Yoshimatsu S, Yamashita Y, 50. Pacella CM, Bizzarri G, Magnolfi F, et al. Laser
Matsukawa T, Takahashi M, Sagara K. Treatment thermal ablation in the treatment of small hepa-
of hepatocellular carcinoma: value of percuta- tocellular carcinoma: results in 74 patients. Radi-
neous microwave coagulation. AJR 1995;164: ology 2001;221(3):712-720.
1159-1164. 51. Cooper IS. Cryogenic surgery: a new method of
39. Matsukawa T, Yamashita Y, Arakawa A, et al. destruction or extirpation of benign or malig-
Percutaneous microwave coagulation therapy nant tissues. N Engl J Med 1963;268:743-749.
in liver tumors: a 3-year experience. Acta Radiol 52. Kane RA. Ultrasound-guided hepatic cryo-
1997;38:410-415. surgery for tumor ablation. Semin Intervent
40. Yamanaka N, Tanaka T, Oriyama T, Furukawa K, RadioI1993;10:132-142.
Tanaka W, Okamoto E. Microwave coagu- 53. Mala T, Edwin B, Tillung T, Kristian Hoi P,
lonecrotic therapy for hepatocellular carcinoma. Soreide 0, Gladhaung 1. Percutaneous cryoabla-
World J Surg 1996;20:1076-1081. tion of colorectal liver metastases: potentiated
41. Dong BW, Liang P, Yu XL, et al. Sonographically by two consecutive freeze-thaw cycles Cryobiol-
guided microwave coagulation treatment of liver ogy 2003;46(1):99-102.
cancer: an experimental and clinical study. AJR 54. Group d'Etude et de Traitment de Carcinome
1998;171:449-454. Hepatocellulaire. A comparison of lipiodol
42. Amin Z, Brown SG, Lees WR. Liver tumor chemoembolization and conservative treatment
ablation by interstitial laser photocoagula- for unresectable hepatocellular carcinoma.
tion: review of experimental and clinical N Engl J Med 1995;332;1256-1261.
studies. Semin Intervent Radiol 1993;10:88- 55. Pelletier G, Ducreux M, Gay P. Treatment of
100. unresectable hepatocellular carcinoma with lipi-
43. Hashimoto D, Takami M, Idezuki Y. In depth odol chemoembolization: a multicenter random-
radiation therapy by YAG laser for malignant ized trial. J HepatoI1998;29:129-134.
tumors in the liver under ultrasonic imaging. 56. Bruix J, Llovet JM, Castelis A, et al. Transarte-
Gastroenterology 1985;88:1663. rial embolization versus symptomatic treatment
44. Steger AC, Lees WR, Walmsley K, Brown Sa. in patients with advanced hepatocellular carci-
Interstitial laser hyperthermia: a new approach noma. Results of a randomised controlled trial in
to local destruction of tumours. BMJ 1989; a single institution. Hepatology 1998;27:1578-
299:362-365. 1583.
45. Nolsoe CP, Torp-Pedersen S, Burcharth F, et al. 57. Caturelli E, Siena D, Fusilli S, et al. Transcatheter
Interstitial laser hyperthermia of colorectalliver arterial chemoembolization for hepatocellular
metastases with a US-guided Nd-YAG laser with carcinoma in patients with cirrhosis: evaluation
a diffuser tip: a pilot clinical study. Radiology of damage to nontumorous liver tissue. Radiol-
1993;187:333-337. ogy 2000;215:123-128.
46. Vogi TJ, Muller PK, Hammerstingl R, et al. 58. Llovet JM, Real MI, Montana X, for the
Malignant liver tumors treated with MR Barcellona Clinic Liver Cancer Group. Arterial
imaging-guided laser-induced thermotherapy: embolization or chemoembolization versus
technique and prospective results. Radiology symptomatic treatment in patients with unre-
1995;196:256-257. sectable hepatocellular carcinoma: a randomised
47. Amin Z, Bown SG, Lees WR. Local treatment of controlled trial. Lancet 2000;359:1734-1739.
colorectal metastases: a comparison of intersti- 59. Matusani S, Sasak Y, Imaoka S, et al. The assess-
tial laser photocoagulation (ILP) and percuta- ment of preoperative transcatheter arterial
neous alcohol injection (PAl). Clin Radio11993; embolization for hepatocellular carcinoma:
48:166-177. the comparison between "whole-liver" TAE and
25. Ablation for Hepatocellular Carcinoma 331

"lobar or segmental" TAE (in Japanese) a mean follow-up of more than 1 year. AIR
Nippon Shokakibyo Gakkai Zasshi 1991;88: 2000;175:1619-1625.
2757-2762. 64. Murakami R, Yoshimatsu S, Yamashita Y, Sagsra
60. Llovet JM, Bruix 1. Systematic review of ran- K, Arakawa A, Takahashi M. Transcatheter
domised trials for unresectable hepatocellular hepatic subsegmental arterial chemoemboliza-
carcinoma: chemoembolization improves sur- tion therapy using iodized oil for small hepato-
vival. Hepatology 2003;37(2):429-442. cellular carcinoma. Correlation between lipiodol
61. Meloni MF, Goldberg SN, Livraghi T, et al. accumulation pattern and local recurrence. Acta
Hepatocellular carcinoma treated with radio Radiol 1994;35:576-580.
frequency ablation: comparison of pulse inver- 65. Bruix J, Castells A, Bosch J, et al. Surgical resec-
sion contrast-enhanced harmonic sonography, tion of hepatocellular carcinoma in cirrhotic
contrast-enhanced power-Doppler sonography patients: prognostic value of perioperative portal
and helical CT. AJR 2001;177(2):375-380. pressure. Gastroenterology 1996;3:1018-1022.
62. Solbiati L, Martegani A, Leen E, Correas JM, 66. Noun R, Jagot P, Farges 0, Sauvanet A, Belghiti
Burns PN, Becker D. Contrast-Enhanced Ultra- 1. High preoperative serum alanine transferase
sound of Liver Disease. New York: Springer, levels: effect on the risk of liver resection in
Verlag Italia, 2003. pp 103-113. Child grade A cirrhotic patients. World J Surg
63. De Baere T, Elias D, Dromain C, et al. Radiofre- 1997;21:390-395.
quency ablation of 100 hepatic metastases with
26
Radiofrequency Ablation of
Neuroendocrine Metastases
Thomas D. Atwell, 1. William Charboneau, David M. Nagorney,
and Florencia G. Que

Neuroendocrine tumors encompass a wide This chapter reviews carcinoid and islet cell
spectrum of neoplasms with a common origin tumors. Different treatment options tradition-
in the neuroendocrine cell system. They share ally incorporated into the management of
the capacity for hormone secretion that results hepatic metastases from these tumors include
in distinct clinical syndromes. Although neuro- surgery and embolization. Radiofrequency
endocrine tumors can arise in organs such as ablation of neuroendocrine hepatic metastases
the adenohypophysis, thymus, lung, thyroid, is reviewed, including a brief review of the
adrenal medulla/paraganglia, and skin (1), most literature.
familiar are the gastroenteropancreatic neuro-
endocrine tumors that include carcinoid and
islet cell neoplasms. Given the high occurrence
of hepatic metastases within this subgroup of Carcinoid Tumor
neuroendocrine tumors, they are a model for
the management of other hormonally active Carcinoid tumors are slow-growing neoplasms
tumors. that arise primarily from three sites in the gas-
Gastroenteropancreatic neuroendocrine neo- trointestinal tract: the appendix, rectum, and
plasms, which include the carcinoid and islet small intestine. Small-bowel carcinoids are the
cell tumors, are a rare group of neoplasms, with most common (3), and probably the most sig-
an incidence of one to two cases per 100,000 nificant clinically because of the symptoms of
people (2). They are relatively unique in that bowel ischemia that occur with mesenteric
they typically are associated with an indolent spread. In as many as 40% of patients, the car-
rate of growth, and frequently are associated cinoid tumor is discovered incidentally during
with tumor-specific hormone production and a the evaluation of an unrelated clinical
secondary clinical syndrome. condition (4).
Because these neoplasms have a unique Metastatic disease is present in 45% of
pathophysiology, the treatment approach is carcinoid patients at the time of diagnosis (3).
different from the traditional management of These tumors are apt to metastasize to the liver
hepatic metastases. Much of what we know over extended time, with metastases eventually
about aggressive management has been learned occurring in up to 85% of patients (5). This slow
through surgical experience, with interven- rate of spread allows the liver to adapt to the
tional radiology providing insight into more bulky metastatic disease. Patients with both
palliative therapy. Radiofrequency ablation carcinoid and islet cell hepatic metastases clas-
(RFA) will likely prove itself to be a valuable sically present with a markedly enlarged liver,
treatment modality for neuroendocrine hepatic yet these patients are fully functional with
metastases. minimal symptoms (6).

332
26. Radiofrequency Ablation of Neuroendocrine Metastases 333

Carcinoid syndrome is a unique symptomatic gross functional tumor burden. Despite the
manifestation of advanced carcinoid disease, frequency of hepatic metastases, these tumors
characterized by flushing, diarrhea, asthma, and often behave in a protracted manner. In
bronchospasm, and, in late disease, tricuspid patients with metastatic disease to the liver, the
valve regurgitation. Carcinoid syndrome is 3-year survival is 56% and the overall survival
encountered in about 7% to 10% of all carci- rate is 42% (8). Death is often secondary to
noid patients, and in 35% to 50% of patients hepatic insufficiency related to tumor burden
with hepatic metastases (4,7). The syndrome is or complications from hormone production.
typically associated with elevated levels of
urine 5-hydroxyindoleacetic acid (5-HIAA).
Carcinoid tumors are notorious for having Treatment of Neuroendocrine
a slow, indolent disease course. Historically,
surgery has been the standard of treatment, Hepatic Metastases
providing prolonged disease-free survival and
cure in many patients. Among patients with The frequency of metastatic disease, the
limited disease treated for curative intent, 80% hormone production, and the protracted
five-year survival has been achieved, although clinical course make gastroenteropancreatic
only 23% of the patients remained recurrence neuroendocrine hepatic metastases responsive
free at twenty-five years (6). Even with incur- to multiple therapeutic modalities that include
able metastatic disease to the liver, median sur- surgical resection, hepatic artery embolization,
vival time exceeds 3 years, and 5-year survival cryotherapy, and radiofrequency thermal abla-
approaches 21 % to 30% (6-8). tion. The surgical experience has formed the
foundation for the management of neuroen-
docrine hepatic metastases.

Islet Cell Neoplasms of


the Pancreas Surgery
The standard of care for patients with
Pancreatic islet cell neoplasms are derived from neuroendocrine cancer metastatic to the liver
neuroendocrine cells. Slightly greater than 50% is now resection of the primary tumor mass
of these tumors are considered functional and hepatic metastases when technically feasi-
because they produce various hormones (8). ble. While bulky and often numerous, these
Such tumors often are named for the charac- metastases often are circumscribed, and they
teristic hormone produced; these include insulin- cause a simple mass effect on adjacent liver
oma, gastrinoma, glucagonoma, VIPoma, and parenchyma, as opposed to invasion or encase-
so on. Occasionally, these tumors produce more ment of adjacent structures. This allows for
than one hormone, which results in a mixed excision with optimal margins.
clinical picture or a change in the hormone syn- Patients considered for surgery include those
drome over time. As with carcinoid and other with preoperative imaging studies that suggest
neuroendocrine tumors, production of a spe- resectability of primary and regional tumor, as
cific hormone leads to characteristic syndromes well as resectability of 90% or more of the
that often result in substantial morbidity and hepatic tumor volume (10). Such treatment has
mortality. resulted in increased survival of patients, up to
Hepatic metastases frequently occur in 73% to 93% at 4 years (10,11) and 61 % to 73%
patients who have pancreatic islet cell neo- at 5 years (12,13). Survival at 10 years is 35%,
plasms, eventually developing in up to 5% to which emphasizes the indolent, yet relentless
10% with insulinoma, 23% to 90% with gastri- nature of this malignancy. There is no appre-
noma, and 70% to 75% with glucagonoma (9). ciable difference in 5-year survival between
The unique hormonal syndrome associated patients with carcinoid tumors and patients
with each tumor typically is proportional to the with islet cell tumors (13).
334 T.n. Atwell et al

Perhaps more significant is the effect of Hepatic Artery Embolization


resection on the control of symptoms. The
degree of endocrinopathy is proportional to Hepatic artery embolization, with or without
tumor volume. Thus, symptom control gained combined intraarterial infusion of chemothera-
by debulking of hepatic metastases is signifi- peutic agents, is an effective alternative to
cant. Relief of hormone-related symptoms surgical resection in patients with surgically
occurs in 96% of patients following resection unresectable metastases. This therapy makes
(13). Complete relief of symptoms is achieved use of the exclusive hepatic arterial supply of
in 86% of patients with carcinoid syndrome neuroendocrine metastases compared to the
treated with palliative intent (14). The mean normal liver parenchyma, which receives nearly
duration of symptom relief following resection 75% of its blood supply from the portal vein.
of neuroendocrine metastases to the liver is 45 After establishing the hepatic arterial anatomy,
months, with recurrence of symptoms in 59% of either the right or left hepatic artery is selec-
patients at 5 years (13). Complete response is tively embolized using particles, absorbable
more prolonged when hepatic resection is per- gelatin, or iodinated oil. Only one lobe is
formed for cure. embolized during a session to avoid hepatic
We have found no significant difference in insufficiency.
the survival rates or initial symptomatic Both isolated particle embolization and
response between patients who had curative chemoembolization have proven effective in
resection and patients who had palliative or treating neuroendocrine hepatic metastases. In
incomplete resection of hepatic metastases a study of isolated particle embolization, the
from neuroendocrine cancer (10). With this in response rate was 96% with duration ranging
mind, the goal of cytoreductive surgery has from 6 to 18 months, depending on the clinical
evolved into either complete resection of the scenario (17). Other investigators have had less
tumor mass or resection of greater than 90% of success with mid-gut carcinoid tumors, report-
the tumor burden. ing a response rate of 52% and median dura-
Despite these promising results of curative tion of effect of 12 months (18). These same
resection and cytoreduction, surgical resection investigators reported a 50% response rate in
of these metastases is possible in only 10% of patients with islet cell metastases to the liver
patients (15). Bilobar liver disease is present in with the median duration of 10 months. With
most patients (76%), and in approximately chemoembolization, the expected response rate
50% of the patients the tumor involves more is approximately 87% and the expected dura-
than 50% of the liver (13,16). In addition, the tion is 11 months (5).
required surgery is often quite extensive and In a study comparing patients who had
associated with significant perioperative risk. hepatic artery occlusion with and without sub-
Major hepatectomy (excision of more than one sequent systemic chemotherapy, the patients
lobe) may be required in more than half of the who also received subsequent chemotherapy
patients (13). In a review of 74 patients by Que had a greater degree of objective tumor regres-
et al (10), more than half of the patients had sion (80% compared to 60%), and a more pro-
major or extended hepatic resections and con- longed response (18.0 months compared with
current intestinal and pancreatic resections. 4.0 months) (19).
Perioperative mortality from such resections is
1.2% to 2.7% (13).
Cryotherapy
Consistent with the natural history of neuro- Cryotherapy has been incorporated in the
endocrine malignancy, recurrence of disease treatment of neuroendocrine hepatic metas-
occurs in a large majority of these patients. tases (20-22). Cryoablation systems deliver
Recurrence following resection occurs in 84 % liquid nitrogen or argon gas to the tip of a thin
of patients at 5 years and 94% at 10 years (13). probe after it has been placed into the metas-
It is very difficult to cure patients of this tasis, commonly under ultrasound guidance. A
disease. series of freeze-thaw cycles is performed,
26. Radiofrequency Ablation of Neuroendocrine Metastases 335

destroying the tumor. Cryotherapy is effective neuroendocrine hepatic metastases: (1) In our
in alleviating symptoms and reducing tumor experience, the most frequent application for
markers in more than 90% of patients (20-22). RFA is the debulking of hepatic metastases to
In a study by Seifert et al (20), 52 neuroen- palliate symptoms and increase survival (Fig.
docrine metastases in 13 patients were treated 26.1). (2) Treatment with RFA may be used
with intraoperative cryotherapy. Five of the with the goal of cure in patients who have a
seven symptomatic patients had complete alle- limited number of hepatic metastases.
viation of symptoms; the remaining two had Currently, specific indications for RFA in
partial relief. A significant reduction in levels of treating neuroendocrine hepatic metastases
serum tumor markers also was observed among are for patients who: (1) need intraoperative
patients for whom data were available. Mor- ablation as an adjunct to resection (Fig. 26.2),
bidity occurred in four of the 13 patients: (2) have limited hepatic disease, but are not
coagulopathy necessitated reoperation in two operative candidates, (3) are inoperable and
carcinoid patients, and acute renal failure and unresponsive to embolization treatments, or (4)
pulmonary embolism occurred in one patient have recurrence after resection (Fig. 26.3).
each. Dedicated studies of RFA of neuroendocrine
Bilchik et al (22) summarized their results for hepatic metastases are limited.The largest study
using intraoperative cryotherapy to palliate to date reported results from using laparoscopic
disease in 19 symptomatic patients with neuro- RFA to treat 34 patients with 234 neuroen-
endocrine metastases to the liver. Estimated docrine hepatic metastases (23). Relief of symp-
tumor destruction during the surgery was 70% toms was achieved in 95% of the patients,
to 90% of the total tumor bulk. Several patients including complete relief in 63%. Mean dura-
had concurrent resection of the primary tumor tion of response was 10 months. Data from 32
or hepatic metastases. All the patients had a of the patients showed local recurrence at 3%
transient coagulopathy that was treated with of the treated metastases, documented between
fresh frozen plasma or platelets. All 19 patients 6 and 12 months following RFA. New liver
were symptom free following surgery, and 18 of metastases developed in 28% of the patients at
the 19 had a reduction in tumor markers. a mean follow-up of 1.6 years. After the treat-
Cozzi et al (21) described six patients who ment, 65 % of the patients had a decrease in the
had hepatic metastases from neuroendocrine level of at least one hormonal marker. Two com-
tumors treated with intraoperative cryother- plications occurred, including transient atrial
apy. All six patients were treated successfully fibrillation and a hepatic abscess, the latter
with no local recurrence at median follow-up of treated by percutaneous drainage.
24 months. The four symptomatic patients had Hellman et al (24) reported 21 patients who
complete relief of symptoms. Levels of tumor had 43 neuroendocrine hepatic metastases
markers decreased by more than 89% in the treated with either percutaneous or intra-
three patients with positive markers. Coagu- operative RFA. Fifteen of these patients were
lopathy requiring additional surgery occurred treated with curative intent, that is, an attempt
in two of the three patients with carcinoid. was made to ablate all lesions. The authors used
a cooled-tip electrode to treat lesions that
ranged in size from 2 to 7 cm (mean, 2.0 cm). At
Radiofrequency Ablation follow-up (mean, 2.1 years), 41 lesions had no
evidence of local recurrence; this resulted in a
Perhaps the most important lesson from the success rate of 95%. Local recurrence devel-
surgical literature is that both complete and oped in only two treated lesions, each at 6
near-complete excision of hepatic metastases months following treatment. Absence of resid-
can have a significant impact on both patient ual disease was documented in only four of the
symptoms and survival. Therefore, RFA can 15 patients treated with curative intent. There
be expected to accomplish two objectives in were two non-life-threatening complications.
the management of gastroenteropancreatic The authors also showed that monitoring levels
336 T.D. Atwell et al

A B C

D E F

G H

Figure 26.1. Radiofrequency ablation (RFA) of symptom control. Contrast-enhanced computed


metastatic glucagonoma causing symptomatic ery- tomography (CT) performed hours after the RFA
thema. A 67-year-old woman who had prior wedge shows large corresponding ablation defects in the
resections of hepatic metastases, presented with liver parenchyma (D,E). The patient’s rash resolved
worsening symptomatic rash, compatible with char- within days of the RFA. At the 2-month follow-up,
acteristic necrolytic migratory erythema (A), and the patient was asymptomatic, the rash had healed
elevated serum glucagon of 16,000 pg/mL. Magnetic (F), and the serum glucagon was 3700 pg/mL. Con-
resonance imaging showed multiple hepatic metas- trast-enhanced CT at this follow-up visit showed that
tases, with three dominant lesions (B,C). Percuta- the bulk of tumor remained destroyed with only
neous ultrasound-guided RFA was performed on small nodular recurrences (arrows) at the ablation
these three largest metastases for debulking and sites (G,H).
A B

c o

E F
FIGURE 26.2. Intraoperative RFA of metas- cial metastases were resected and intraop-
tatic functional pheochromocytoma. A 22- erative ultrasound-guided RFA was per-
year-old woman presented with a history formed on the two deeper lesions (D). CT
of pheochromocytoma and worsening pal- obtained 3 days following ablation showed
pitations and hypertension that required both the surgical wedge defects (E) and the
therapy with multiple antihypertensive RFA ablation defects (F). The patient's
medications. Magnetic resonance imaging hypertension resolved, and she was dis-
(MRI) showed recurrence of tumor in the charged from the hospital without antihy-
left adrenal bed and four hepatic metastases pertensive medication.
(arrows) (A-C). At operation, the superfi-
338 T.D. Atwell et al

A B C

D E F

G
Figure 26.3. Radiofrequency ablation (RFA) of metastatic carcinoid. A
66-year-old man, 1 year after liver debulking surgery, presented with
diarrhea, elevated levels of urine 5-hydroxyindoleacetic acid (5-HIAA)
(80 mg/24 hours), and multiple hepatic metastases (A,B). Percutaneous
ultrasound-guided RFA of the two largest hepatic metastases was per-
formed for tumor debulking and control of symptoms (C). Contrast-
enhanced CT performed immediately following RFA showed large
corresponding ablation defects in the liver (D,E). The patient’s diarrhea
improved following RFA and urine 5-HIAA decreased to 39 mg/24
hours. Contrast-enhanced CT performed 9 months following RFA
showed complete local control, evident by reduction of the ablation zone
and absence of enhancement (F,G).

of tumor markers after treatment is difficult therapy was decreased, and tumor size was
due to the presence of disease elsewhere in the smaller on follow-up computed tomography
abdomen. (CT) scans. There were no complications.
Wessels and Schell (25) reported the use of Results of these studies show that RFA
intraoperative RFA in three patients for treat- relieves symptoms in patients who have
ment of carcinoid hepatic metastases refractory neuroendocrine tumors. Although the cure of
to hepatic artery embolization (24). All three neuroendocrine malignancy is challenging,
patients suffered from malignant carcinoid RFA provides a relatively safe method of tumor
syndrome. In all three, symptoms improved debulking, and the rate of local recurrence
after RFA treatment, the dosage of octreotide is low.
26. Radiofrequency Ablation of Neuroendocrine Metastases 339

Summary 5. Proye C. Natural history of liver metastasis of


gastroenteropancreatic neuroendocrine tumors:
place for chemoembolization. World J Surg 2001;
Neuroendocrine malignancies are rare neo-
25:685-688.
plasms frequently associated with characteristic 6. Moertel CG. Karnofsky memorial lecture. An
hormonal syndromes. Gastroenteropancreatic odyssey in the land of small tumors. J Clin Oncol
neuroendocrine tumors frequently develop 1987;5:1502-1522.
hepatic metastases. Their biologic behavior 7. Moertel CG, Sauer WG, Dockerty MB,
affords multiple treatment options. If surgically Baggenstoss AH. Life history of the carcinoid
feasible, resection of metastases is the best tumor of the small intestine. Cancer 1961;14:901-
option for the patient. Hepatic artery emboliza- 912.
tion is also effective in nonsurgical patients, and 8. Thompson GB, van Heerden JA, Grant CS,
provides at least short-term palliation. Carney JA, Ilstrup DM. Islet cell carcinomas of
Radiofrequency ablation is evolving into an the pancreas: a twenty-year experience. Surgery
1988;104:1011-1017.
effective method of tumor destruction, and it
9. Carty SE, Jensen RT, Norton JA. Prospective
can improve patient symptoms. As demon- study of aggressive resection of metastatic
strated in the surgical literature, removal of the pancreatic endocrine tumors. Surgery 1992;112:
bulk of neuroendocrine hepatic metastases 1024-1031; discussion 1031-1032.
improves patient survival. On the basis of this 10. Que FG, Nagorney DM, Batts K~ Linz LJ, Kvols
surgical experience, we expect a similar effect LK. Hepatic resection for metastatic neuro-
on patient survival with RFA. endocrine carcinomas. Am J Surg 1995;169:36-
Probably the most significant contribution of 42; discussion 42-43.
RFA will be as adjunctive treatment intra- 11. Grazi GL, Cescon M, Pierangeli F, et al. Highly
operatively and postoperatively. Coordination aggressive policy of hepatic resections for neuro-
of surgical and ablation techniques during the endocrine liver metastases. Hepatogastroen-
terology 2000;47:481-486.
course of surgery allows optimal treatment of
12. Chen H, Hardacre JM, Uzar A, Cameron JL,
the neuroendocrine malignancy through surgi- Choti MA. Isolated liver metastases from neuro-
cal excision of both the primary tumor and endocrine tumors: does resection prolong sur-
accessible hepatic metastases, as well as con- vival? J Am ColI Surg 1998;187:88-92; discussion
current ablation of deep-seated lesions in the 92-93.
liver. This affords the surgical option to a larger 13. Sarmiento JM, Heywood G, Rubin J, Ilstrup DM,
percentage of patients who have metastases Nagorney DM, Que FG. Surgical treatment of
that were previously deemed unresectable. Fol- neuroendocrine metastases to the liver: a plea
lowing surgery, RFA allows minimally invasive for resection to increase survival. Journal Amer
treatment of the almost inevitable recurrence ColI Surgeons 2003;197(1):29-37.
of disease in the remaining liver. 14. Que FG, Heywood GM. Hepatic surgery for
metastatic gastrointestinal neuroendocrine
tumors. In: Blumgart LH, Fong Y, Jarnagin W,
References eds. Hepatobiliary Cancer. American Cancer
Society and B.C. Dekker, 2000;97-116.
1. DeLellis RA, Tischler AS. The dispersed neuro- 15. McEntee GP, Nagorney DM, Kvols LK, Moertel
endocrine system. In: Kovacs K, Asa SL, eds. CG, Grant CS. Cytoreductive hepatic surgery
Functional Endocrine Pathology. Malden, MA: for neuroendocrine tumors. Surgery 1990;108:
Blackwell Science, 1998:529-549. 1091-1096.
2. Oberg K. State of the art and future prospects in 16. Chamberlain RS, Canes D, Brown KT, et al.
the management of neuroendocrine tumors. Q J Hepatic neuroendocrine metastases: does inter-
Nucl Med 2000;44:3-12. vention alter outcomes? J Am ColI Surg 2000;
3. Modlin 1M, Sandor A. An analysis of 8305 cases 190:432-445.
of carcinoid tumors. Cancer 1997;79:813-829. 17. Brown KT, Koh BY, Brody LA, et al. Particle
4. Shebani KO, Souba WW, Finkelstein DM, et al. embolization of hepatic neuroendocrine metas-
Prognosis and survival in patients with gastroin- tases for control of pain and hormonal symp-
testinal tract carcinoid tumors. Ann Surg 1999; toms. J Vasc Intervent Radiol 1999;10:397-
229:815-821; discussion 822-823. 403.
340 T.n. Atwell et al

18. Eriksson BK, Larsson EG, Skogseid BM, 22. Bilchik AJ, Sarantou T, Foshag U, Giuliano AE,
Lofberg AM, Lorelius LE, Oberg KE. Liver Ramming KP. Cryosurgical palliation of metas-
embolizations of patients with malignant tatic neuroendocrine tumors resistant to conven-
neuroendocrine gastrointestinal tumors. Cancer tional therapy. Surgery 1997;122:1040-1047;
1998;83:2293-2301. discussion 1047-1048.
19. Moertel CG, Johnson CM, McKusick MA, et al. 23. Berber E, Flesher N, Siperstein AE. Laparo-
The management of patients with advanced scopic radiofrequency ablation of neuro-
carcinoid tumors and islet cell carcinomas. Ann endocrine liver metastases. World J Surg 2002;
Intern Med 1994;120:302-309. 26:985-990.
20. Seifert JK, Cozzi PJ, Morris DL. Cryotherapy 24. Hellman P, Ladjevardi S, Skogseid B, Akerstrom
for neuroendocrine liver metastases. Semin Surg G, Elvin A. Radiofrequency tissue ablation using
OncoI1988;14:175-183. cooled tip for liver metastases of endocrine
21. Cozzi PJ, Englund R, Morris DL. Cryotherapy tumors. World J Surg 2001;26:1052-1056.
treatment of patients with hepatic metastases 25. Wessels FJ, Schell SR. Radiofrequency ablation
from neuroendocrine tumors. Cancer 1995;76: treatment of refractory carcinoid hepatic metas-
501-509. tases. J Surg Res 2001:95:8-12.
27
Tumor Ablation in the Kidney
Debra A. Gervais and Peter R. Mueller

The interventional radiologist who establishes tion to the details specific to ablation of RCC.
a percutaneous tumor ablation program will This chapter reviews the current clinical man-
likely become skilled in liver tumor ablation agement of RCC, and the clinical and technical
before attempting to treat renal cell carcinoma. considerations in performing RFA for renal
Liver tumors are more common than primary tumors. In addition, postablation imaging find-
renal tumors, and most patients with liver ings are reviewed, along with the associated
tumors are not surgical candidates. Thus, a challenges inherent in imaging follow-up.
steady influx of patients eligible for percuta-
neous ablation of hepatic tumors can be Renal Cell Carcinoma: Staging,
expected in many centers. In contrast, surgical
removal remains the standard therapy for small
Prognosis, and Current Therapies
renal cell carcinomas, and most patients can
undergo resection (1). Nevertheless, with the
Staging and Prognosis
incidence of renal cell carcinoma (RCC) Staging systems for RCC have undergone
increasing and with the aging population in the several revisions over the years. Each revision
United States, the number of patients with represents an attempt to improve the correla-
small RCCs who are not good surgical candi- tion of staging with prognosis. In 1997 the
dates is expected to increase (2-4). Thus, from American Joint Committee on Cancer estab-
time to time, the radiologist experienced in lished the current staging system for RCC that
percutaneous tumor ablation can expect to is summarized in Table 27.1 (5). As we shall see,
encounter patients for whom percutaneous the limitations of tumor size for which there is
treatment of RCC is the only option or the a reasonable chance to achieve local control
most reasonable one. with RFA is well within 7 cm, the size criterion
In current practice, the least expensive, most that the American Joint Committee on Cancer
readily available, and most straightforward per- (AJCC) staging system designates as the
cutaneous therapeutic option is radiofrequency demarcation between stage I and stage II for
ablation (RFA). Most RFA educational sym- RCCs that have not spread beyond the kidney
posia or syllabi are devoted predominantly to (5-7). Thus, the radiologist who performs RFA
liver ablation with all of its nuances. Renal of RCC will be focused predominantly on stage
tumors, if they are discussed at all, are often I tumors. While the search for the perfect prog-
included under the rubric "extrahepatic" abla- nostic tool continues, tumor staging remains the
tion. This overarching categorization includes most reliable and thus the most important para-
ablation of tumors of the lung, head and neck, meter (8).
adrenals, spleen, and miscellaneous abdominal Other tumor features have been studied for
and pelvic tumors, thereby leaving little atten- their prognostic utility. Among the most famil-

341
342 D.A. Gervais and P.R. Mueller

TABLE 27.1. Renal cell carcinoma TNM classifica- system (9). However, the prognostic utility has
tion and staging system of the American Joint proven variable when evaluated by different
Committee on Cancer (AJCC). investigators. In addition, grading can be sub-
TNM Definitions jective among pathologists. For these reasons,
Primary tumor (T) grading remains of secondary importance to
Tl Tumor 7 cm or less, limited to kidney staging for practical clinical applications.
T2 Tumor more than 7 cm, limited to kidney
T3 Tumor extends into major veins or invades
With respect to histology, there are four types
adrenal or perinephric tissues, but not of renal cell carcinoma (10). The most common
beyond Gerota's fascia type is clear cell carcinoma, also known as con-
T3a Tumor invades adrenal or perinephric tissues, ventional RCC. Clear cell carcinoma accounts
not beyond Gerota's fascia for approximately 70% of solid renal masses in
T3b Thmor grossly extends into renal vein(s) or
Ive below diaphragm surgical series. The next most common histo-
T3c Tumor grossly extends into renal vein(s) or logic type of RCC is papillary, or chromophil,
Ive above diaphragm RCC, which accounts for 10% to 15% of solid
T4 Tumor invades beyond Gerota's fascia renal masses. Some investigators have reported
a more favorable prognosis than for clear cell,
Regional lymph nodes (N)
NX Nodes cannot be assessed
whereas other investigators have shown a worse
NO No lymph nodes prognosis. The prognosis of papillary RCC com-
Nl Metastasis in a single regional node pared to clear cell RCC remains controversial
N2 Metastases in more than one regional node and further studies regarding cytogenetics and
molecular markers may clarify this issue.
Distant metastases (M)
MX Metastases cannot be assessed
Rarer RCCs are the chromophobe type (5%)
MO No metastases present with a favorable prognosis compared to clear
Ml Metastases present cell RCC (10). On the other hand, collecting
duct RCCs «1 %) are aggressive and metasta-
Staging size early. In large part, the remainder of renal
Stage I Tl,NO.MO
masses are tumors that cannot be subtyped into
Stage II T2, NO, MO one of these four categories, either because of
Stage III Tl, Nl, MO features of more than one type or due to sar-
T2,Nl,MO comatoid changes. Sarcomatoid change is not a
T3a,NO,MO unique subtype, as it can be seen in any of the
T3a. Nl,MO
T3b, NO,MO
underlying histologic types (10). However, it
T3b,Nl,MO carries a universally poor prognosis, regardless
T3c,NO,MO of the underlying histologic type of RCC. Other
T3c,Nl, MO terminology that may be encountered in the
Stage IV T4, NO, MO clinical evaluation of renal masses includes
T4,Nl,MO
AnyT,N2,MO
papillary adenoma, metanephric adenoma, and
Any T, Any N, Ml oncocytoma. These are generally considered
benign tumors.
Note: For T3 tumors, adrenal involvement refers to direct Finally, there are several new promising
invasion only, not to hematogenous metastatic spread.
prognostic features that currently are being
IVe, inferior vena cava.
evaluated; these include cytogenetic and molec-
ular markers (11). For the time being, these
remain experimental and have yet to enter
iar are grading and tumor histology (9,10). widespread clinical use.
Grading of renal cell carcinoma is based on
nuclear characteristics, whereas histology is
based on tissue architecture. In North America,
Current Therapy
the Fuhrman grading system, which consists of The treatment for RCC in the absence of distant
four grades, is the most commonly utilized metastases is surgical resection (1). The conven-
27. Tumor Ablation in the Kidney 343

tional procedure has been radical nephrectomy, rounded by perinephric fat, are favorable for
a procedure that carries a 1% to 2% mortality RFA. The fat serves as an insulator that facili-
risk and 15% to 30% morbidity (1,12,13). tates maintenance of high temperatures within
Although it may not always be feasible, the tumors, thereby enhancing necrosis (6,7).
nephron-sparing surgery (partial nephrectomy) This phenomenon has been termed the oven
allows for preservation of renal function and has effect by Livraghi et al (16), who described
proven equally effective. Developments in improved coagulation necrosis in encapsulated,
laparoscopic resection have reduced recovery and hence insulated, hepatomas. On the other
times for patients in whom the this method hand, central tumors with extensions into the
is appropriate (1). Nevertheless, despite the renal sinus are more difficult to treat with RFA
diminished morbidity and mortality associated (6,7). This increased difficulty is related to per-
with these less invasive options, an even less fusion-mediated tissue cooling, the heat-sink
invasive procedure may be indicated in selected effect, mediated by the large central blood
patients such as those with existing comorbid vessels (17). The constant inflow of blood at
conditions that make anesthesia or surgery body temperature results in cooling of the por-
highly risky, those patients with multiple tumors tions of tumor adjacent to large blood vessels.
such as occur in von Hippel-Lindau (VHL) This limits both the peak temperatures and the
disease, or the elderly. These patients might tol- duration of these temperatures in regions of
erate a percutaneous procedure better than a tumor that as a consequence may undergo
laparoscopic resection, and they are the impetus inadequate coagulation necrosis (17).
for new clinical developments in tumor ablation. Thus, the ideal tumor for percutaneous RFA
is a 3 cm or less exophytic tumor without direct
contiguity to vital structures such as bowel or
Indications/Contraindications for ureter (7). Tumors up to 5 cm are also reason-
Percutaneous Radiofrequency able candidates. Small central tumors (those
with a component in the renal sinus) can
Ablation undergo successful RFA, but proximity to large
renal arteries or veins is almost guaranteed as
Ideal Tumor
the tumor size enlarges. The cooling effect of
The limits on the volume of coagulation necro- the large vessels may preclude successful abla-
sis that can be achieved with a single applica- tion, or may result in the need for additional
tion of RFA and the basic science underlying ablation sessions to achieve complete tumor
this limitation are discussed elsewhere in this necrosis (7).
volume. Based on experimental observations, Finally, when assessing a tumor for possible
one can reasonably expect to achieve complete RFA, staging relies on imaging and clinical
coagulation necrosis of most RCCs that are evaluation. Although not a common occur-
3cm or less (14). In addition, one can reason- rence, metastases can occur even with small
ably expect to achieve complete necrosis of primary RCCs. Thus, review of an abdominal
tumors up to 5 cm, but some of these may computed tomography (CT) or magnetic reso-
require more ablation sessions (6,14). These nance imaging (MRI) exam of a renal mass for
hypothetical expectations have been confirmed potential RFA includes assessment of the
by initial clinical reports of RFA of RCC (6). retroperitoneal lymph nodes, inferior vena
While absolute size limits are difficult to estab- cava, liver, lung bases, bones, and contralateral
lish, one is likely to encounter a rapid decline kidney for metastases or synchronous tumors.
in rates of complete coagulation necrosis as
tumor size increases beyond 5 cm. This has been
confirmed for liver tumors and by initial results
Patient Selection
for kidney tumors (7,15). Despite promising results following percuta-
Tumor location is also likely to influence neous RFA of RCCs with up to 4 years of
RFA success. Exophytic tumors, partially sur- follow-up available in a few patients, RFA
344 D.A. Gervais and p.R. Mueller

remains a relatively new procedure to treat unsafe due to proximity to adjacent ureter or
stage I RCC (6,7,18,19). In the treatment of bowel, laparoscopic mobilization of these adja-
RCC, the historical standard to which new tech- cent structures may facilitate ablation if the
niques have been compared is open radical patient is a suitable candidate for anesthesia
nephrectomy (1). Newer less invasive proce- and if the tumor cannot be removed. The
dures such as partial nephrectomy and laparo- optimal technique in these situations is to
scopic nephrectomy have been required to combine the laparoscopic expertise of the urol-
demonstrate similar rates of 5-year survival and ogist with that of image-guided ablation of the
disease-free survival for the results to be con- radiologist. Finally, in the unlikely event of a
sidered equivalent. Using this benchmark, complication that requires retrograde ureteral
partial nephrectomy has proven as effective as stent placement, a urologist familiar with the
complete nephrectomy, and initial reports on 5- patient is most appropriate.
year results following laparoscopic resection
likewise show results similar to open resection Planning Radiofrequency
(1). The currently available clinical experience
with RFA of RCC is limited to several series Ablation of Renal Cell
of 25 to 50 patients with mean postablation Carcinoma
periods of approximately 1 to 1.5 years
(6,7,18,19). Based on the tumor and patient criteria defined
Until long-term data are available, we are above, an algorithm for triaging patients can be
limiting RFA for the treatment of RCC to implemented. The first of two critical questions
select patients. The primary patients are those to answer is this: Is the patient a good candidate
who are unable to undergo nephrectomy for surgical resection based on the evaluation
because of comorbid diseases that make the of a urologist? If so, then, proceed to resection.
surgical risk of morbidity and mortality exces- If not, the second question follows: Is percuta-
sively high. Typically, these comorbid diseases neous image-guided RFA technically safe and
include cardiovascular or pulmonary condi- feasible? Review of available imaging is
tions as well as other malignancies (6). In addi- mandatory to begin to answer this question.
tion, patients with less than a 10-year life These first two steps allow rapid screening of
expectancy based on comorbid conditions or patients who are not suitable for RFA, either
advanced age are reasonable candidates for because they can have surgery or because the
percutaneous ablation (6). We do require that tumor size, location, or proximity to other struc-
there be a reasonable likelihood of at least 1 tures makes RFA impossible or unsafe.
year of life expectancy, as any patient with a Once a stage I RCC is deemed suitable for
condition with expected mortality within a year percutaneous RFA, the next step is to assess the
is not likely to experience any deleterious effect adequacy of the available imaging. The role of
on health from a stage I RCC. Patients for imaging for RFA is not only to screen the case
whom surgical resection would risk or guaran- for suitability, but also four other important
tee a dependency on dialysis also are candi- functions:
dates for RFA. These include patients with
• To choose a modality for procedure guidance
solitary kidneys and patients with predisposi-
• To define preablation margins-the whole
tion to multiple RCCs, such as occur in VHL
target for RFA
disease or in familial cases of RCC (6,7,18).
• To serve as a baseline for postablation
Finally, in selecting patients for percutaneous
imaging
RFA, we have found collaboration with a urol-
• To plan the ablation approach-percuta-
ogist to be indispensable (6). The involvement
neous or laparoscopic
of the urologist ensures thorough assessment
of all therapeutic options for the patient. In If any of these functions is not adequately
patients for whom resection may be difficult served by available imaging or if imaging is not
and for whom percutaneous ablation is deemed recent, then additional cross-sectional imaging
27. Throor Ablation in the Kidney 345

should be performed to address all of these may not be safe. However, if the patient is a
issues. At a minimum, a contrast-enhanced suitable candidate for anesthesia, such as in a
CT or MRI is needed before RFA since these relatively young patient with VHL disease and
are the imaging modalities that will be used multiple RCCs, then laparoscopic mobilization
to monitor the result. Ideally, the modality of these structures may allow RFA. If laparo-
that is used for postablation imaging is avail- scopic ablation is performed, imaging guidance
able at baseline to facilitate comparison. In in the form of operating room ultrasound still
current practice, CT remains the modality of is needed to ensure optimal electrode position-
choice; MRI is used only for patients who ing for complete tumor necrosis.
cannot receive intravenous (IV) contrast
material because of renal insufficiency or prior
anaphylaxis. Preablation Biopsy
The choice of imaging modality to guide RFA
is limited in clinical practice to ultrasound or The role of needle biopsy of renal masses
CT. When compared to CT guidance, ultra- remains controversial and is evolving. Standard
sound is generally more readily available and urology practice is to remove solid renal masses
allows real-time assessment of the needle tip without biopsy (1). For most patients with solid
position that facilitates and expedites needle renal masses, this approach is not likely to
adjustments. However, depending on body change soon. However, there are many patients
habitus and tumor location, ultrasound visual- for whom a biopsy diagnosis may be desirable
ization of a given tumor may be suboptimal to prior to surgery or other invasive therapy. For
guide ablation. Furthermore, while ultrasound example, in the setting of another primary
may demonstrate a portion of the RCC, all of malignancy, active or in remission, biopsy may
the tumor must be well-visualized with ultra- be prudent (1,21). Wood et al (21) found in his
sound to allow accurate needle and electrode series that 12 of 24 patients with primary malig-
placement for overlapping (or even single) nancies other than RCC had solid renal masses
ablations to achieve complete coagulation that were metastases, not RCC. Furthermore,
necrosis. If the tumor cannot be assessed com- management changed in 32 of 79 (41 %) cases
pletely with ultrasound, then CT may be a based on biopsy results.
better choice for guidance. Magnetic resonance In patients with another primary malignancy,
guidance has shown promising results, with biopsy should be considered before proceeding
clear advantages of both accurate intraproce- with RFA. In some patients, biopsy may not be
dural monitoring of the necrosis margins and necessary. For example, Pavlovich et al (18) did
imaging guidance of the needle electrode in off- not perform preablation renal mass biopsy
axial planes of imaging (20). However, in North in their series of 24 tumors since all patients
America, there are no commercially available had VHL disease or hereditary RCC. In the
MR-compatible RF generators and needle elec- Pavlovich series, all masses were enhancing
trode systems. Thus, at present MR guidance on CT and had shown growth on serial CT.
remains in the realm of researchers with dedi- Radiofrequency ablation of an enhancing
cated equipment. enlarging solid renal mass is reasonable regard-
Accurate delineation of the tumor extent is less of the results of the needle biopsy, and we
critical, both for achieving complete coagula- have occasionally performed ablation of such a
tion necrosis when performing RFA and for mass in the setting of a nondiagnostic or nega-
serving as a baseline comparison for postabla- tive needle biopsy. However, our practice is to
tion imaging. Tumor margins can be obscured perform preablation biopsy since urologists at
by the kidney if available CT is not contrast our institution manage biopsy-proven oncocy-
enhanced or if the timing of the contrast bolus tomas as benign neoplasms that are followed
is suboptimal for renal mass evaluation. closely by imaging, and not resected in older
Finally, in cases of RCC in direct contiguity patients. This particular management of onco-
with bowel or ureter, percutaneous ablation cytoma is not universal practice, as there is a
346 D.A. Gervais and P.R. Mueller

divergence of opinion among urologists on this longer analgesic action. We are currently giving
issue (1). ondansetron (4 mg IV) as an antiemetic as
We have adopted the practice pattern of our needed.
local urologists and do not perform RFA of
oncocytomas. Thus, needle biopsy prior to
ablation serves to triage solid renal masses.
Optimizing Ablation: Choice of
Because there remains regional variability in
Systems and Performing
the approach to oncocytomas, the radiologist
Overlapping Ablations
undertaking RFA of renal masses should be Since the major focus of preclinical and early
familiar with the preferred management of clinical work in RFA of soft tissue tumors has
local urologists and plan accordingly. been the liver, generator design and ablation
protocols for commercially available systems
for soft tissue tumor ablation have been opti-
mized for ablation of liver tumors. Experi-
Performing Radiofrequency mental preclinical work on RFA for renal
Ablation of Renal tumors is more limited, and in practice, most
radiologists use the same ablation parameters
Cell Carcinoma for renal tumors as prescribed for liver tumors
(6,7,23,24). The kidney is similar to the liver in
Sedation
that it too is a very vascular parenchymal organ.
Most patients are able to tolerate percutaneous Nevertheless, the electrical and thermal char-
RFA as an outpatient procedure performed acteristics are likely different from liver, given
with IV sedation (6,7). Since the duration ofthe the different electrolyte milieu. Critics of rapid
procedure is longer than for a typical biopsy or assimilation of RFA into clinical use for extra-
drainage procedure, the recovery from the IV hepatic soft tissue tumors have suggested that
sedation also can be slightly longer. For these the use of system parameters optimized for
reasons, RFA is best performed early enough in liver tumors may not be optimal for the treat-
the day to allow sufficient time for the proce- ment of renal tumors. While further experi-
dure and recovery the same day. Medically mental work may shed new insights into the
complex patients may require overnight obser- features unique to the renal response to
vation following RFA (7). In some cases, the thermal ablation, these features mayor may
patient is admitted the day prior to the proce- not translate into the need for changes in abla-
dure to allow adequate hydration, to achieve tion protocols. Given the gross similarities
blood pressure control, and/or to correct a between the two organs, the results of early
coagulopathy if needed. Rare patients may clinical experience and the current knowledge
require assistance of an anesthesiologist who regarding renal response to thermal ablation,
may administer sedation or general anesthesia the current clinical approach appears justified
as appropriate. (6,7,18,19).
Intravenous sedation regimens vary among The available RF generator systems and their
institutions, and must be kept within estab- compatible needle electrodes are reviewed in
lished institutional guidelines (22). In our prac- detail elsewhere in the book. Each available
tice, we have achieved adequate sedation and generator system has prescribed end points
analgesia in almost all cases with a combination for terminating a given ablation using one of
of midazolam (2-5mg), fentanyl (100-300mg), three parameters: rapid rise in impedance
and meperidine (25-50mg) (6,7). Our initial (Radiotherapeutics, Boston Scientific, Natick,
cases received droperidol as the third agent MA), target temperature (RITA, Mountain
both for its antiemetic and calming effects, but View, CA), or time (Radionics, Burlington,
droperidol is no longer available (6). Thus, we MA).1f an umbrella array electrode is chosen,
have added meperidine for most patients for its the diameter should be similar to the tumor
27. Tumor Ablation in the Kidney 347

size. If the tumor is larger than the largest avail- For small tumors that require a single abla-
able electrode, then overlapping ablations will tion, the needle electrode must be positioned as
be needed. The straight needle electrodes come near to perfectly central as possible so as to
in single or clustered arrangements (Radionics) avoid peripheral residual viable tumor. If this
with electrically active tips of 2.0 and 3.0cm for central positioning is not readily achieved and
the single electrodes and 2.5 cm for the cluster if an eccentric ablation is performed, then addi-
electrodes. For small tumors up to 2.5 cm, a tional ablations can be performed to compen-
single 2- or 3-cm active tip is adequate. For sate for any anticipated regions of viable tumor.
tumors 2.5 cm or larger, a cluster electrode In reality, multiple overlapping ablations are
provides more rapid necrosis of the tumor, needed for most tumors based not only on size,
although one can achieve coagulation necrosis but also on tumor geometry. Even if needle
of masses 2.5 to 4.0cm with multiple overlap- electrodes could produce perfectly predictable
ping ablations with a single electrode. and reproducible thermal lesions, tumors do
Tumor ablation requires more meticulous not come in perfect spheres or cylinders. They
technique than needle biopsy. Biopsy requires may have an oval shape or irregular projections
needle placement within a tumor. Once diag- that necessitate special attention to achieve
nostic cells are obtained, the procedure is over. complete tumor necrosis. Whereas a simple
Needle repositioning is needed only if rapid eccentric projection of a tumor can be antici-
cytologic or frozen-section evaluation fails to pated to require a dedicated eccentric needle
identify diagnostic material. Tumor ablation, electrode placement and ablation, the more dif-
however, requires induction of cell death ficult issue is how to predict optimal needle
throughout the entire three-dimensional electrode placement to achieve thermal lesions
volume of the target tumor (25). This task that completely overlap a 4- to 5-cm tumor
requires strategic needle electrode placement (Figs. 27.1 and 27.2).
whether for a single ablation or for multiple Computed tomography and ultrasound find-
overlapping ablations (14,25). In theory, ings do not predict the margins of necrosis
umbrella array systems create thermal lesions accurately during RFA. Thus, the positioning of
in the shape of spheres, and perfectly over- electrodes in performing overlapping ablations
lapped ablation spheres create a larger com- is made by the operator, based on size and
posite thermal lesion with the narrowest geometry of the tumor, expected size and shape
diameter at the surface indentations of the of the thermal lesions from the electrode
three-dimensional composite (25). In theory, system in use, and any anticipated effects from
straight needle electrodes create cylindrical tumor surroundings such as perfusion-medi-
thermal lesions of varying diameter, depen- ated tissue cooling from large vessels at any
dent on single or cluster arrangement, and edge of the tumor (7). In addition, tissue tem-
slightly longer than the electrically active shaft perature may provide some insight into
(14). whether the thermal lesion is at the larger or
In practice, however, a different picture smaller end of the range that can be achieved.
usually emerges. The reality is that the shapes For internally cooled systems, the ideal tem-
of the thermal lesions can be irregular, of vari- perature response following cessation of
able diameters, and difficult to predict. Fur- cooling and of electrical current at 12 minutes
thermore, the diameters of overlapping thermal is a rapid rise in temperature to well over 60°C
lesions created by the same needle electrode and slow return to less than 60°C. A tempera-
during the same session and in the same tumor ture less than 60°C will achieve coagulation
may vary. Thus, decision making during RFA necrosis in a smaller volume of tissue. For
regarding positioning of electrodes is based in the temperature controlled system (RITA), the
part on empirical science, but a large part of the computer chip alerts the operator to check the
practical application of RF ablation of soft temperature at 30 seconds after termination of
tissue tumors remains an art. the ablation. A rapid decrease in temperature
A B
FIGURE 27.1. Computed tomography (CT) scan in an (B) CT scan following IV contrast material again
81-year-old man with a solitary kidney and a 4.2-cm shows the RCC now with a better determination of
biopsy-proven renal cell carcinoma (RCC) (arrow). the medial margin. The RCC enhanced to 139
(A) CT scan without IV contrast material shows an Hounsfield units.
exophytic RCC that measures 48 Hounsfield units.

A B
FIGURE 27.2. CT scan during and immediately following
radiofrequency ablation (RFA). (A) CT scan with the
patient prone shows cluster needle electrode (curved arrow)
near the medial margin of the tumor. Note that a small
amount of hemorrhage (small arrow) displaces the tumor
from the colon. (B) CT scan shows the cluster electrode
(arrow) repositioned into the lateral and inferior aspects of
the tumor. Again seen is a small amount of hemorrhage
(small arrow) anteriorly. The electrode handle (arrowhead)
is demonstrated. When positioning the patient for CT-
guided RFA, the operator must leave enough room for the
patient and the electrode handle to fit within the CT gantry.
(C) CT scan performed after final removal of the electrodes
and a total of four overlapping ablations shows hemorrhage
anterior and posterior to the kidney (small arrows). There is
scattered air adjacent to the tumor. Note the linear bands of
increased density (arrow) within the tumor. This finding is
c thought to be from desiccation related to RFA.
27. Thmor Ablation in the Kidney 349

indicates fast tissue cooling and suggests the sions to achieve complete necrosis (7).Although
thermal lesion will not be at the large end of multiple sessions are less than ideal, for some
the range. If rapid cooling is encountered, the patients who are unable to have surgery, a
umbrella device can be redeployed in the exact second session is not unreasonably onerous if it
same location following rotation of the tines so can achieve complete tumor necrosis.
as to deploy them in different locations within
the tumor. The straight needle system can be
repositioned more closely than might be the Complications and Postablation
case if high temperatures are achieved and
maintained. In addition, once a straight needle Syndrome
is repositioned, the temperature reading over
60°C suggests that it is in an area already The most commonly reported major complica-
treated by the prior ablation. Although no tion associated with percutaneous RFA of RCC
single parameter allows accurate prediction of is hemorrhage (6,7,18,19). In all reported cases,
the final ablation margins, with experience the hemorrhage and its treatment resulted
operator can integrate all of this information in no prolonged deleterious outcomes. Other
and perform overlapping ablations accordingly reported complications include ureteral stric-
(Fig. 27.2). tures (7). Although not yet reported, injury
For exophytic tumors, some authors have to other organs or structures such as bowel,
suggested that the first ablation be strategically adrenal gland, spleen, lung, or pancreas is pos-
performed at the interface between tumor and sible. Skin burns at the grounding pad sites also
normal kidney so as to limit blood flow to the can occur. To date, there have been no reports
remainder of the tumor and thereby facilitate of procedure-related mortality.
coagulation necrosis for the remaining overlap- The postablation syndrome, described as flu-
ping ablations (26). When easily achievable, this like symptoms, consisting of generalized aches,
is a reasonable approach. However, depend- malaise, and fevers to 103°F can occur after
ing on the location and orientation of the RFA, although we have not seen this syndrome
tumor-kidney interface, percutaneous place- as commonly with RCC as with liver tumors.
ment of a straight needle electrode directly at Although anecdotally felt to be more common
the interface may not be possible due to inter- following larger volume ablations, the inci-
vening structures. Indeed, tumors have been dence of this syndrome and the predisposing
treated successfully with RFA in our experi- factors are not well known, especially with
ence with a needle electrode approach perpen- respect to ablation of RCC.
dicular to the tumor-kidney interface.
Ablation sessions are terminated once the
operator has performed overlapping ablations Postablation Imaging
from which complete coagulation necrosis is
anticipated (7). For very large tumors, more Since necrotic tumor is left in situ, imaging
than one session can be anticipated. The remains the only effective means of evaluation
maximum ablation that can be achieved in any of the entire tumor volume, and thus plays a
session usually is based on the duration of ade- critical role in the evaluation of patients fol-
quate analgesia and sedation determined by lowing RFA. The key determinant of residual
limits set by institutional protocol (7). viable tumor following RFA ablation is the
Ideally, for patient convenience and for cost uptake of contrast material administered intra-
minimization, a strategy that achieves complete venously (6,7,18,27). Portions of tumor that do
tumor treatment in a single session is desirable. not enhance reflect regions of coagulation
Tumors 3 cm or smaller are almost all treated necrosis (Fig. 27.3). Regions that demonstrate
with a single session. However, for tumors 3 cm persistent enhancement reflect residual disease.
or larger, a significant minority of patients in our This interpretation of imaging findings is
experience has required second ablation ses- extrapolated from radiologic-pathologic corre-
350 D.A. Gervais and P.R. Mueller

A B

FIGURE 27.3. CT scan 1 week after ablation. (A) CT scan


without IV contrast material shows stable hemorrhage
(small arrows) and tumor (large arrows). The tumor now
measured 60 Hounsfield unites. (B) CT following IV con-
trast material shows stable hemorrhage (small arrow) and
tumor (large arrow) that measured 62 Hounsfield units.
(C) CT during excretory phase shows hemorrhage (small
arrows) and tumor (large arrows). Tumor remained at 62
Hounsfield units. Following RFA, the entire tumor must be
evaluated for evidence of enhancement that would suggest
c viable tumor.

lation work performed in hepatic tumors (27). may be seen following RFA (18,28). Although
Goldberg et al (27) were able to confirm coag- CT is the modality used for assessment of RFA
ulation necrosis that corresponded to areas of in most patients, for patients who cannot
nonenhancement to within 2mm. Persistent receive IV contrast material because of renal
peripheral nodular enhancement correlated insufficiency or prior anaphylactoid reaction,
pathologically with viable tumor. Ongoing clin- MRI, without and with gadolinium-chelate
ical experience supports the use of this CT enhancement may be useful (6,7).
interpretation scheme for postablation imaging Imaging generally is performed at more fre-
of renal tumors. quent intervals in the early postablation period.
On postablation CT, the tumor typically does Generally, we have been assessing patients
not disappear. A decrease in maximum diame- within approximately 1 month, 3 months, and 6
ter by 1 to 5mm is common (7,18). Nonen- months (6,7). Once the I-year CT is reached,
hancing regions of adjacent renal parenchyma and no viable tumor has been demonstrated,
27. Thmor Ablation in the Kidney 351

then CT scans can be performed at I-year isolated metastatic RCC that appear after
intervals. nephrectomy. Reports of successful treatment
of isolated lung nodules and lymph nodes have
already appeared (30,31).
Clinical Experience to Date
While only a few reports have appeared in the Conclusions
literature, the delays from manuscript prepara-
tion to publication guarantee a larger cumula- Radiofrequency ablation of kidney tumors has
tive experience than the approximately 90 cases shown encouraging early results using modern
reported thus far. The total cumulative experi- RF-generator systems designed for soft tissue
ence of most of the large centers performing tumor ablation. While 5-year data are pending,
RFA of RCC is now likely to be well over many limitation of this therapeutic option to patients
hundreds of patients. The reported series who are not good candidates for surgical
provide strong support for the use of RFA for resection of RCC is prudent. Postablation
small tumors, less than 3.0 cm. For exophytic imaging is critical for assessment of ablation
tumors up to 5.0cm, our experience has been success, and radiologists must be familiar with
that up to 30% of these patients may require a the appearance of necrotic tissue and viable
second ablation session, but complete coagula- tumor.
tion necrosis can be achieved in all cases (7).
Review of published series must take into References
account changes in technology that evolved
1. Novick AC, Campbell Sc. Renal tumors. In:
rapidly over the middle to late 1990s. Genera- Walsh PC, ed. Campbell's Urology. 8th ed.
tor systems have become more powerful so that Philadelphia: WB Saunders, 2002:2672-2731.
early preclinical or clinical suboptimal results 2. Greenlee RT, Murray T, Bolden S, Wingo PA.
need to be reevaluated with the more powerful Cancer statistics 2000. CA Cancer J Clin 2000;
modern systems. Several early reports were 50:7-33.
based on work with 50-W generators. An early 3. Jayson M, Sanders H. Increased incidence of
study that compared the three available serendipitously discovered renal cell carcinoma.
systems found that the most powerful created Urology 1998;51:203-205.
the largest lesion, suggesting that power output 4. Chow WH, Devesa SS, Warren JL, Fraumeni JE
might be a factor that limited the size of Rising incidence of renal cell cancer in the
United States. JAMA 1999;281:1628-1631.
thermal lesions with less strong generators (29).
5. Kidney. In: American Joint Committee on
With most generator systems now in clinical use Cancer: AJCC Cancer Staging Manual. 5th ed.
in the 150- to 250-W range, further increases in Philadelphia: Lippincott-Raven, 1997:231-234.
power are not likely to further augment the 6. Gervais DA, McGovern FJ, Wood BJ, Goldberg
volume of a single ablation. SN, McDougal WS, Mueller PRo Radio-fre-
Several developments in RFA of RCC are quency ablation of renal cell carcinoma: early
likely in the foreseeable future. Radiofre- clinical experience. Radiology 2000;217:665-
quency ablation systems are readily available 672.
and relatively inexpensive. Thus, in the near 7. Gervais DA, McGovern FJ, Arellano RS,
future, the use of RFA ablation to treat RCC is McDougal WS, Mueller PRo Radiofrequency
likely to continue to grow as the technology ablation of renal cell carcinoma: clinical experi-
ence and technical success in 42 tumors.
continues to disseminate. To emphasize a point
Radiology 2003;226(2):417--424.
made previously, a more thorough evaluation 8. Gettman MT, Blute ML. Update on pathologic
of outcomes compared to surgical resection can staging of renal cell carcinoma. Urology 2002;
be performed once 5-year results are available. 60:209-217.
These data are likely to be available in the next 9. Medeiros D, Jones EC, Aizawa S, et al. Grading
2 to 3 years. In addition, RFA may be useful in of renal cell carcinoma. Cancer 1997;80:990-
a limited number of patients with small foci of 991.
352 D.A. Gervais and P.R. Miieller

10. Storkel S, Eble IN, Adlakha K, et a1. Classifica- 22. American Society of Anesthesiologists Task
tion of renal cell carcinoma. Cancer 1997;80: Force on Sedation and Analgesia by Non-
987-989. Anesthesiologists. Practice guidelines for seda-
11. Swanson DA, Rothenberg HJ, Boynton AL, et a1. tion and analgesia by nonanesthesiologists.
Future prognostic factors for renal cell carci- Anesthesiology 1996;84:459-470.
noma. Cancer 1997;80:997-998. 23. Zlotta AR, Wildschutz T, Raviv G, et a1.
12. Uzzo RG, Novick AC. Nephron sparing surgery Radiofrequency interstitial tumor ablation
for renal tumors: indications, techniques and out- (RITA) is a possible new modality for treatment
comes. J Urol2oo1;166:6-18. of renal cancer: ex vivo and in vivo experience.
13. Reddan DN, Raj GV, Polascik TJ. Management J EndouroI1997;11:251-258.
of small renal tumors: an overview. Am J Med 24. Zlotta AR, Schulman Cc. Ablation of renal
2001;110:558-562. tumors in a rabbit model with interstitial saline-
14. Goldberg SN, Gazelle GS, Mueller PRo Thermal augmented radiofrequency energy. Urology
ablation therapy for focal malignancy: a unified 1999;54:382-383.
approach to underlying principles, techniques, 25. Dodd GD III, Frank MS, Aribandi M, Chopra S,
and diagnostic imaging guidance. AJR 2000;174: Chintapalli KN. Radiofrequency thermal abla-
323-331. tion: computer analysis of the size of the thermal
15. Livraghi T, Goldberg SN, Lazzaroni S, et a1. injury created by overlapping ablations. AJR
Hepatocellular carcinoma: radiofrequency abla- 2001;77: 777-782.
tion of medium and large lesions. Radiology 26. Dupuy DE, Mayo-Smith WW, Cronan JJ. Image-
2000;214:761-768. guided biopsy and radiofrequency ablation of
16. Livraghi T, Goldberg SN, Lazzaroni S, Meloni F, renal masses. Semin Intervent Radiol 2000;17:
Solbiati L, Gazelle GS. Small hepatocellular car- 373-380.
cinoma: treatment with radiofrequency ablation 27. Goldberg SN, Gazelle GS, Compton CC, Mueller
versus ethanol injection. Radiology 1999;210: PR, Tanabe KK. Treatment of intrahepatic
655-661. malignancy with radiofrequency ablation:
17. Goldberg SN, Hahn PF, Tanabe KK, et a1. Per- radiologic-pathologic correlation. Cancer 2000;
cutaneous radiofrequency tissue ablation: does 88:2452-2463.
perfusion mediated tissue cooling limit coagula- 28. Gervais DA, O'Neill MJ, Arellano RS,
tion necrosis? J Vase Intervent Radiol 1998;9: McGovern FJ, McDougal WS, Mueller PRo Peri-
101-111. tumoral CT changes associated with radio-
18. Pavlovich CP, Walther MM, Choyke PL, et a1. frequency (RF) ablation of focal renal lesions:
Percutaneous radio frequency ablation of small description, incidence, and significance.
renal tumors: initial results. J Urol 2002;167: Radiology 2001;221(P):180.
10-15. 29. de Baere T, Denys A, Kuoch V, Elias D, Vilgrain
19. Mayo-Smith WW, Dupuy DE, Ridlen MS, V, Roche A. Radio-frequency tissue ablation of
Cronan JJ. Radiofrequency ablation of solid the liver: in vivo and ex vivo experiments with
renal masses: results of 26 consecutive treat- four different systems. Eur Radiol 2003;13(10):
ments in 22 patients. AJR 2003;180(6):1503- 2346-2352.
1508. 30. Zagoria RJ, Chen MY, Kavanagh PV, Torti FM.
20. Lewin JS, Connell CF, Duerk JL, et a1. Interac- Radiofrequency ablation of lung metastases
tive MRI-guided radiofrequency interstitial from renal cell carcinoma. J Urol 2001;166:
thermal ablation of abdominal tumors: clinical 1827-1828.
trial for evaluation of safety and feasibility. 31. Gervais DA, Arellano R, McDougal WS,
J Magn Reson Imaging 1998;8:4D-47. McGovern FJ, Mueller PRo Radiofrequency
21. Wood BJ, Khan MA, McGovern F, Harisinghani (RF) ablation of metastatic soft tissue tumors of
M, Hahn PF, Mueller PRo Imaging guided biopsy the genitourinary system: indications, results, and
of renal masses: indications, accuracy and impact the role of RFA after failed conventional thera-
on clinical management. J Urol 1999;161:1470- pies. Radiology 2001;221(P):261.
1474.
28
Radiofrequency Ablation for
Thoracic Neoplasms
Sapna K. Jain and Damian E. Dupuy

Lung cancer statistics in the United States esti- best current therapies result in an overall 5-
mate that 171,900 people in 2003 were diag- year survival rate, for all stages combined, of
nosed with lung cancer (1).As the leading cause only 15% (1). The poor response of lung cancer
of cancer death among men and women in the to current treatment methods necessitates the
United States, the associated death rate for lung use of alternative modalities.
cancer is 28%, surpassing mortality rates of Less invasive therapies that can accomplish
colon, prostate, and breast cancer combined tumor destruction or complete eradication
(1). This startling fact underscores the impor- without the use of general anesthesia may com-
tance of improved methods to treat this aggres- plement, improve, or replace existing therapies.
sive form of cancer. Its ominous prognosis is One such ablative tumor therapy that may add
reflected by the overall 5-year survival rates to the treatment regimen in this complex group
for previously untreated patients with primary of patients is radiofrequency ablation (RFA).
non-smaIl-cell lung cancer (NSCLC) after sur- Percutaneous image-guided tumor RFA is an
gical treatment (according to pathologic stage): expanding minimally invasive modality for
63% to 67% in stage lA, 46% to 57% in IB, the local treatment of solid malignancies. First
52% to 55% in IIA, 33% to 39% in lIB, and reported in 2000 (5), RF ablation of human
19% to 23% in IliA (2,3). lung tumors may be a promising treatment
With NSCLC, 30% of patients present with option for nonsurgical candidates, given the
disease confined to the parenchyma, 30% with suboptimal outcomes with current treatment.
spread to intrathoracic lymph nodes, and 40% The insertion of an RF electrode into the
with metastatic disease (4). In cases of regional defined tumor bed and establishment of an
disease, a combination of surgery, chemother- electric field to a reference electrode that
apy, and external beam radiation therapy [x-ray oscillates with generated alternating RF cur-
therapy (XRT)] prevails as standard treatment. rents ultimately create a conduit for frictional
For patients with localized disease not invading heating (6). This tissue heating consequently
the mediastinum, surgical resection remains induces coagulative necrosis and cell death in a
the best treatment option. Surgery is not the controlled and predictable manner (6). The sur-
primary treatment when there are coexistent rounding air in the normal parenchyma of the
morbid medical conditions or advanced stage lung acts as an insulator and concentrates RF
of the disease. In these patients who are not sur- energy in the targeted tissue, thereby requiring
gical candidates, the treatment options rely pri- less energy deposition (7). The high vascular
marily on XRT, with or without chemotherapy. flow of the lung results in a "heat-sink" effect
In small-cell lung cancer and in other cases in that dissipates heat away from normal adjacent
which radiotherapy cannot be administered, tissue and concentrates the effective energy
chemotherapy may be administered solely. The deposition within the solid component of the

353
354 S.K. Jain and D.E. Dupuy

lesion. This chapter discusses the clinical ratio- nonoperative treatments, but its useis becom-
nale, technique, potential applications, and ing more widely accepted (10,11). Pulmonary
early clinical experience in thoracic RFA. metastasectomy for isolated pulmonary metas-
tases of different tumor types results in an
overall 5-year survival rate of approximately
Clinical Scenarios 36% (12), and is associated with a mortality
rate of <2% (11). Indications for surgery
General include exclusion of other distant disease, no
tumor at the primary site, probability of com-
Lung neoplasms are a heterogeneous group of plete resection, and adequate cardiopulmonary
tumors. Primary lung cancer can be categorized reserve to withstand the operation. However,
into several different cell types: squamous cell there are several clinical scenarios in which
carcinoma, adenocarcinoma (including bron- local therapy via surgery or alternative treat-
choalveolar), large cell carcinoma, and small ment modalities may improve survival or
cell carcinoma. These different cell types have provide palliation when these indications are
different biologic properties; thus the disease not met. Cited 5-year survival rates range are
presentation and treatment may differ accord- as follows for pulmonary metastasectomy:
ingly. For example, the effective response rate osteogenic sarcoma, 20% to 50%; soft tissue
to XRT in cases of lung cancer metastatic to sarcoma, 18% to 28%; head and neck carci-
bone varies with cell type: 80% for small cell noma, 40.9% to 47%; colon carcinoma, 21 % to
carcinoma, 72% for adenocarcinoma, and 40% 48% (13), breast carcinoma, 31 % to 49.5% (11);
for squamous cell carcinoma (8). As stated and renal cell carcinoma, 20% to 44% (14).
earlier, stage at presentation is a critical deter- Repeat resection is supported in select patients
minant of the treatment strategy. Stage IV who are free of disease at the primary location,
(metastatic disease) is quite variable in its but have recurrent metastatic disease to the
clinical course and outcome. Certain metastatic lung (15,16).
histologies such as osteosarcoma tend to be
isolated to the lungs, have an indolent growth
pattern similar to renal cell carcinoma or colo- Palliative Care
rectal carcinoma (9), and therefore are more Given that many patients with lung neoplasms
likely to respond to a local form of therapy. present with advanced disease and that the
New minimally invasive treatments must majority of patients with lung cancer (86%)
bring new benefits for patients. A shotgun will die from their disease (17,18), one can
approach could be detrimental to the patient, deduce that at least one of the following
as well as hinder research and acceptance of symptoms/problems will clinically manifest
any new treatment modality. As with the appli- itself during the course of the disease: pain,
cation of any new treatment, utilization of a dyspnea, cough, hemoptysis, metastases to the
multidisciplinary team ensures a prudent clini- central nervous system or musculoskeletal
cal approach with the patient's interests para- system, tracheoesophageal fistula, or obstruc-
mount. Pulmonary tumor boards or general tion of the superior vena cava (18). Symp-
oncology tumor boards are excellent venues for tomatic palliation, therefore, becomes an
educational exchange and should be attended important part of treatment, yet the current
by anyone engaged in the treatment of lung medical literature reports failure to do so (19),
neoplasms. with 50% of patients dying without adequate
pain relief (20). Approximately 5% of lung
Metastatic Disease cancers extend beyond the lung to invade the
pleura, soft tissues, or osseous structures of the
Surgical treatment of metastatic lung cancer is chest wall (21).
controversial, given the lack of randomized Three main causes of malignancy-related
controlled clinical trials comparing surgery with pain in lung cancer are osseous metastatic
28. Radiofrequency Ablation for Thoracic Neoplasms 355

disease (34%), Pancoast tumor (31%), and vasculature, and RFA may be considered
chest wall disease (21 %) (22). Mechanisms that salvage methods by providing, at minimum, pal-
result in pain include tumor progression and liative relief to patients who fail conventional
related pathology (e.g., periosteal inflamma- modalities. Utilization of RFA in the treatment
tion, nerve damage), side effects of radiation or of pain due to trigeminal neuralgia and osteoid
chemotherapy, infection, and muscle aches sec- osteoma has been documented (29,30).
ondary to limited physical capability. Approxi-
mately 70% to 90% of patients with advanced
cancer have pain, and effective palliation can be
Chest Wall Invasion
achieved in 90% (18,19). Cited 5-year survival rates for NSCLC with
The management of cancer pain is addressed chest wall invasion treated surgically range
by the clinical practical guidelines from the from 15% to 38% (31). When microscopically
Agency for Health Care Policy and Research incomplete or gross residual disease after
(AHCPR) (23, now not current according to its attempted resection is present, overall 5-year
Web site, but referenced in the literature). survival rate declines to 4% (21). Survival rates
Various strategies and potential disadvantages are dependent on the extent of nodal involve-
include analgesic medications (nonsteroidals, ment, depth of invasion, and the completeness
acetaminophen, morphine) that may impair of resection (21). Incomplete resections, even if
mental function; palliative radiation that may leaving only microscopic disease, offer no
fail to affect insensitive tumors, damage normal chance for cure for the patient (21).
tissue, and/or be limited by maximum tolerated
doses; antineoplastic therapies (with the goal of
reducing tumor burden); nerve blocks that have Technique
short-lived effects; and palliative invasive
surgery. Patients referred for lung tumor RFA initially
Conventional treatment of osseous metasta- are evaluated in a clinic setting where the
tic disease involves XRT and chemotherapy. patient history and pertinent imaging studies
X-ray therapy has been shown to palliate are reviewed. At this time, the appropriateness
respiratory symptoms and improve quality of for RFA, risks and benefits of the procedure,
life in patients with NSCLC (24). Current rec- and any additional preprocedural studies are
ommendations by the AHCPR for bone metas- discussed and planned. This preprocedural
tases include pharmacologic use to control evaluation is similar to a surgical evaluation,
pain, followed by XRT to control localized whereby any possible risks of bleeding or
pain. The use of systemic corticosteroids serious cardiopulmonary issues are addressed.
(prednisone, 20-40mg/d), bisphosphonates, Side effects from this procedure, such as post-
calcitonin, and radiopharmaceuticals (e.g., procedural pyrexia secondary to conductive
ethylenediamine tetramethylene phosphonic local heating of tissue and circulatory systemic
acid or strontium-89 chloride) may be used with heating through the bloodstream (32) also are
or without XRT. The results, however, are poor discussed. In general, virtually all patients
to fair, with a small to moderate net benefit. healthy enough to undergo computed tomog-
Furthermore, extension of tumor into the chest raphy (CT)-guided needle biopsy are good can-
wall and osseous metastatic disease can be didates for pulmonary RFA. Patients with only
limited and difficult to treat with XRT, since one lung can safely undergo pulmonary RFA,
prior treatment fields may have encompassed as long as provisions for rapid deployment of a
the symptomatic region. Current treatment chest catheter are made. Patients with severe
thus is often ineffective in complex patients emphysema who require supplemental oxygen
(25-27). Because up to 70% of patients have also can be treated. However, it is important to
disease that is too advanced for resection (28), remember that patients with emphysema who
newer alternatives such as percutaneous retain carbon dioxide may lose their respiratory
ethanol injection, embolization of bone tumor drive when given higher percentages of supple-
356 S.K. Jain and D.E. Dupuy

mental oxygen under conscious sedation during The RF electrode is placed through the skin
an RFA procedure. Patients with underlying and pleura to a length that corresponds to one-
idiopathic pulmonary fibrosis should be con- half to two-thirds the distance to the target
sidered poor candidates for pulmonary RFA, as lesion. A CT-fluoroscopic image is obtained,
exacerbation of the underlying disease may and the RF electrode angle in the x, y, and z
lead to serious respiratory failure and death planes is corrected as necessary.
after the procedure. For pleural-based masses, a shorter RF elec-
To reduce potential complications of trode is used. Placement of the electrode within
sedation-induced nausea and aspiration of the target tumor in this situation may need to
gastric contents, all patients fast overnight. Pa- be performed without initial superficial posi-
tients may take hypertension and cardiac med- tioning, since superficial placement from a
ications in the morning with a small quantity of lateral position may result in protrusion of the
water. Insulin-dependent diabetic patients electrode that limits placement of the patient
should administer half of their usual morning into the CT gantry. A coaxial guiding instru-
insulin dose. An abridged physical exam is per- ment also could be used in this situation,
formed outside the procedure suite and an intra- whereby the RF electrode is placed directly
venous line is placed. Thirty minutes prior to the into the mass after the correct outer cannula
commencement of the procedure, all patients position has been confirmed.
are given 0.625 mg of droperidol intravenously For lesions smaller than 2cm in diameter,
for sedative and antiemetic effects. Patients with central and distal positioning of the RF elec-
Parkinson's disease should not receive droperi- trode usually is adequate for the first ablation
dol, as it may exacerbate symptoms. with subsequent tandem ablation zones per-
Patients then are brought to the CT scanner formed during more proximal positioning.
where the technical staff places the appropriate When at all possible, the target lesion should be
grounding pads on the opposite chest wall from entered along its longitudinal axis to allow for
the skin entry site (e.g., anterior chest wall for this type of sequential overlapping tandem
patient lying prone) to direct the RF current ablations during electrode withdrawal. For
and thus prevent damage to adjacent structures lesions larger than 2 cm in diameter, larger elec-
in the target area. After the initial scout images trodes or multiple overlapping ablation zones
are taken, a skin mark is placed on the patient may need to be performed to ensure adequate
that corresponds to the entry site determined thermocoagulation of the target lesion.
by the computer grid at the appropriate table Ablation of the liver may be used as a com-
position. Horizontal and vertical laser lights in parison to the lung. Similar to liver tumor abla-
the CT gantry correspond to the x- and y-axes tion, working around the periphery of larger
from the computer grid on the screen, and a tumors in the lung helps ensure adequate abla-
ruler can be placed to match the desired skin tion of the soft tissue margins. Ablation of lung
entry site as determined on the computer tumors, in contrast to tumors in the liver, tends
screen. The area is prepped and draped in to require less time and current to achieve ade-
sterile fashion, and local buffered lidocaine quate thermocoagulation. The baseline circuit
anesthesia is administered both intradermally impedance of small parenchymal masses sur-
and to the level of the pleura with a 25-gauge rounded by aerated lung may not allow the
skin needle and a 22-gauge spinal needle, re- same amount of current deposition compared
spectively. Computed tomography fluoroscopy to a liver tumor ablation. Depending on the RF
is initiated, and an image is taken with the equipment employed, each ablation should be
spinal needle in place to identify proper table carried out according to the manufacturer's
position and needle angle. Repositioning can be specifications regarding temperature and/or
performed with the spinal needle if necessary. impedance.
A small skin incision is made at the correct skin Just prior to and during RF heating, patients
entry site by a No. 11 scalpel blade 1 to 2cm are given intravenous conscious sedation with
into the subcutaneous tissues. midazolam (1 mg doses) and fentanyl (50 Ilg
28. Radiofrequency Ablation for Thoracic Neoplasms 357

doses) monitored by continuous vital signs and petroleum jelly-based gauze to provide an air-
an electrocardiogram. The RF heating of small tight seal on removal. In patients with air leaks,
parenchymal masses away from the visceral prolonged chest catheter drainage may be
pleura may require less sedation and may not required, which may include placement of a
produce any pain. On the other hand, pleural- surgical chest tube that will require prolonged
based masses can be quite painful during RF hospitalization. All patients are observed for at
heating. Given the somatic innervation of the least 2 hours postprocedure, and interval
parietal pleura (via the intercostal and phrenic follow-up imaging is scheduled at discharge.
nerves), irritation may result in pain felt either
on the body wall or in the corresponding der-
matomes, for which multiple doses of sedation
during the procedure may be needed. Radio- Potential Applications and Early
frequency heating of central lesions adjacent Clinical Experience
to bronchi elicits a prominent cough response
from patients that may require sedation to The application of radiofrequency energy to
reduce patient motion. Occasionally, general pulmonary neoplasms is a revolutionary
anesthesia may need to be used in pediatric concept whose clinical applications are just
patients or patients who may not tolerate the beginning to be developed. Unlike conven-
RF heating with conscious sedation alone. tional therapies such as chemotherapy and
Dual-lumen endotracheal (ET) tubes are not XRT, multiple applications can be performed
necessary as the pulmonary bleeding from RFA for local control without additional difficulty or
is not any different from that in aCT-guided morbidity. The safety is not significantly differ-
biopsy and may be less, given the coagulating ent from that of a lung biopsy. Almost all of our
effect of the RF current. RF procedures are performed in an outpatient
After the target tumor is treated, the RF setting. Contraindications for RFA are few;
electrode is removed and aCT-fluoroscopic they include uncorrectable bleeding disorders
image is obtained to evaluate for a pneumo- and recent use of anticoagulants. Potential com-
thorax. Large pneumothoraces can be evacu- plications include pneumothorax, hemorrhage,
ated at this time with chest catheters and wall pleural effusion, pleurisy, damage to adjacent
suction. Smaller asymptomatic pneumotho- anatomic structures (low likelihood given the
races can be followed with chest radiographs. In predictable nature of the procedure), skin
the latter group, we put patients on 100% burns secondary to grounding pads, and infec-
oxygen via a nonrebreathing mask and obtain tion. The exact role of RFA for pulmonary neo-
an immediate chest radiograph, followed by a plasms and its complementary applications
2-hour chest x-ray. An increasing pneumotho- with standard therapies remains to be eluci-
rax on this 2-hour follow-up typically necessi- dated. Early data are extremely encouraging,
tates chest catheter placement. Once a chest and continued clinical research will be neces-
catheter has been placed and there is radi- sary to define precisely which patients will
ographic documentation of pneumothorax res- benefit most from this novel therapy.
olution, the patient may be discharged with a The most exciting application for RFA is the
Heimlich valve that is placed on the end of the treatment of primary bronchogenic carcinoma,
catheter. A 24-hour follow-up chest x-ray is specifically NSCLC. With the overallS-year rel-
formed. Hospitalization may be required ative survival rate for all stages less than 15%
because of pain or patient apprehension about (1), newer treatment modalities are sorely
outpatient chest catheter management. If the needed. Since RFA is a local therapy, the clear-
pneumothorax is resolved on follow-up chest x- est indication for its utilization would be in inci-
ray, the tube is checked for an air leak by having dental or screen-detected stage I disease in
the patient cough with the tube end in a con- which the tumor size is less than 3 cm in diam-
tainer of sterile water. If no air bubbles are visu- eter and there is no evidence of regional or
alized, then the tube is removed with a distant spread by staging CT or positron emis-
358 S.K. Jain and D.E. Dupuy

sion tomography (PET) scanning. The gold status. Tumor staging was done by PET scan-
standard therapy for this group of patients is ning, and PET and CT are used to evaluate
lobectomy, which has a 70% five-year survival local progression (Fig. 18.2). Pulmonary toxic-
(33). Studies report overall 5-year survival ity was evaluated by pre- and posttherapy pul-
rates ranging from 53.8% to 82% when both monary function testing. In the 23 patients
anatomic (lobectomy) and nonanatomic treated with this approach with a median
(limited/wedge resection) are included (34-38). follow-up of 16 months, there have been no
However, many patients with stage I tumors are local treatment failures. Four patients have
poor surgical candidates due to severe under- died: one from a cerebrovascular accident 3
lying cardiopulmonary disease. In this group of months after completion of therapy, two from
patients, a limited local therapy such as RFA or early metastatic disease that was detected
radiotherapy may benefit survival compared to within 3 months of RFA, and one patient 18
no treatment (Fig. 28.1). months after therapy secondary to respiratory
In patients with localized disease who can be failure. No significant pulmonary toxicities have
treated safely by RFA, tumor control can be been observed. These early results are encour-
achieved (25). We have just completed a phase aging and we eagerly await the long-term
II trial comparing combination therapy of results.
primary RFA followed by conventional XRT to Pulmonary RFA alone for the treatment of
the known historical data of radiotherapy primary lung cancer is not validated currently,
alone. Given the low sensitivity and specificity given prospective surgical data that demon-
of CT to detect mediastinal lymph nodes, as strate a threefold increase (or 75% increase)
well as the influence of lymph node metastases and 2A-fold increase in the locoregional recur-
on prognosis, PET scanning was used to evalu- rence rate with local treatment (wedge resec-
ate lymph nodes rather than primary tumor tion and segmental resection, respectively)

A c

FIGURE 28.1. A 91-year-old man with biopsy-proven


non-small-cell lung cancer in the left upper lobe.
(A) Supine axial computed tomography (CT) image
showing a 3.0 x 2.0cm mass in the left upper lobe that
was documented to be increasing in size. (B) CT fluo-
roscopy image showing a 2.5-cm active-tip cluster
radiofrequency (RF) electrode within the mass.
(C) Axial CT image at 14-month follow-up shows tumor
B shrinkage with residual scarring.
28. Radiofrequency Ablation for Thoracic Neoplasms 359

Oi

Oii

c Oiii

FIGURE 28.2. A 76-year-old woman with primary prior to RFA (i), 7 months status post-RFA and
non-small-celllung carcinoma (T1NOMO) of the left XRT (ii), and 16 months status posttreatment (iii).
lung. (A) Axial CT showing a circumscribed 2-cm (i) Focal increased area of uptake in the left suprahi-
mass in the mid-aspect of the left upper lobe (arrow). lar region after 6.6mCi of F-18 FDG (arrow). (ii)
(B) Radiofrequency ablation (RFA) of the tumor Previously noted focus of activity in left suprahilar
with the electrode (arrow) in the tumor bed. (C) region no longer seen. iii. Increased uptake in the
Axial CT at 20 months follow-up status post-RFA superficial anterior and posterior thorax (large
and x-ray therapy (XRT) showing tumor contraction arrow) as well as minimum uptake in the left medi-
(large arrow), diffuse emphysematous changes, and astinum (small arrow), likely secondary to radiation-
scarring (small arrows) consistent with XRT. (D) related changes. No evidence of tumor recurrence is
Axial positron emission tomography (PET) images shown.
360 S.K. Jain and D.E. Dupuy

compared to lobectomy (17). Recurrences after tion to recurrence in the current literature (43),
a "complete" resection will develop over the but complications such as radiation pneumoni-
next 5 years in 20% to 30% of patients with tis, esophagitis, or myelopathy are potential
stage I disease, in 50% with stage II, and in 70% hazards.
to 80% with stage III disease (39). More Explanations for recurrence include inade-
current studies that compare limited resection quate resection of the primary tumor and the
(segmentectomy) and lymph node assessment presence of microscopic lymphatic disease
versus lobectomy in patients with stage IA within the lung and ipsilateral hilar nodes.
NSCLC with tumor size less than 2 cm in diam- Unfortunately, these microscopic deposits
eter have shown equivalent rates for overall 5- cannot be detected, and current RF technology
year survival (87.1% vs. 93%) and local does not allow the treatment region to extend
recurrence (40,41). Thus, in a properly selected far enough into normal aerated lung
patient population, wedge resection or seg- parenchyma. Yet for patients who are nonsur-
mentectomy may be superior to nonsurgical gical candidates due to comorbid conditions,
alternatives. Recurrence from residual micro- poor cardiopulmonary reserve, or prior XRT
scopic disease may be delayed or prevented with regrowth in the treatment field, RFA is an
with neoadjuvant and adjuvant chemotherapy alternative (Fig. 28.3). Studies on repeated pul-
and XRT (4,42). External beam reirradiation monary metastasectomy for recurrent disease
also has been documented as a potential solu- have reported 5-year survival rates of 48% (16).

c
A

FIGURE 28.3. A 71-year-old man with non-small-cell


lung carcinoma involving the right upper lobe status
post-chemotherapy and radiation with regrowth within
the radiation field. The patient was not a candidate for
surgery or continued XRT. (A) Axial CT showing a 7.0
x 4.0cm mass (large arrow) in the right upper lobe abut-
ting the pleura. Secondary radiation changes of scarring
and fibrosis are seen adjacent to the mass (small
arrows). (B) CT fluoroscopy image showing RF elec-
trode (arrow) within the tumor bed. (C) Axial CT at
6-month follow-up shows an air-filled cavity with no
B residual soft tissue.
28. Radiofrequency Ablation for Thoracic Neoplasms 361

A B

c D
FIGURE 28.4. A 61-year-old man with metastatic in the tumor periphery (arrows) with the BRT
renal cell carcinoma status post-bilateral pulmonary catheter repositioned posteromedially for further
wedge resection and chemotherapy with new seed placement. (C) Axial CT immediately after
metastatic lesion in the right lower lobe. (A) Prone RFA. Note seeds along the periphery of the tumor
CT axial image shows a 3.5 x 2.5 x 2.0 cm mass bed (arrows). (D) Prone CT axial image at I-year
(arrow) in the medial portion of the right lower lobe. follow-up shows tumor contraction with more
(B) Following RFA, iodine-125 seeds were deposited peripheral seed placement (arrows).

In addition to XRT, utilization of RFA with propensity for infiltrative growth, then tumors
other treatment modalities is increasing. For that tend to be more encapsulated and with
larger asymptomatic parenchymal masses, com- a sharper tumor/lung interface (e.g., well-
bination therapy with brachytherapy implants differentiated squamous cell carcinoma) may
(both high-dose and low-dose radioactive be better candidates for local therapy alone. If
sources) may provide greater local control by concurrent external beam radiotherapy is
magnifying cytoreduction and enhancing the contraindicated, then high-dose radioactive
radiation effect by destruction of centrally (HDR) brachytherapy may provide improved
located hypoxic tumor (25; unpublished data). local control in these patients.
The benefits of concurrent brachytherapy Pulmonary metastatic disease tends to be
with RFA are that the entire treatment can an indicator of widespread systemic disease.
be accomplished in a single day, and the However, in certain tumors and certain pa-
brachytherapy seeds can be placed under CT tients, pulmonary metastatic disease may exist
guidance after the RFA procedure (Fig. 28.4). in isolation. In these patients with a finite
If one analyzes the tumor histologies and their number of metastatic deposits in the lung from
362 S.K. Jain and D.E. Dupuy

a tumor with favorable biologic characteristics detection and staging are essential. In our expe-
(such as soft tissue sarcoma, renal cell carci- rience, localized tumors over 3 cm in size may
noma, colorectal carcinoma, and pulmonary have early, undetected metastatic disease by
carcinoma), resection is considered a viable current imaging and staging techniques. Inva-
treatment option that improves prognosis sive staging techniques (e.g., mediastinoscopy)
(depending on the nodule size, completeness of are not options in these patients with medical
resection, and lymph node status) (44--49). In comorbidities. Therefore, it may be beneficial to
colorectal metastases, for example, studies have have medical oncology expertise early in the
shown the overall 5-year and 10-year survival care of these patients, given the higher likeli-
rates to increase from 22% to 62% and 42% to hood of metastases.
47%, respectively, and a 3-year survival benefit For palliation of symptoms related to a focal
after resection of pulmonary metastases lesion, size is less important, and larger tumors
(47,48). For patients at high risk for morbidity with chest wall and osseous involvement can be
from thoracotomy or for those patients who treated with attention to the tumor/bone inter-
refuse surgery, the only treatment alternatives face (Fig. 28.6). The current literature confirms
are systemic chemotherapy or local therapy the palliative results of RFA in musculoskele-
by external beam radiation. X-ray therapy as tal, gastrointestinal, pulmonary, and neurologic
primary treatment in patients with stage I associated lesions presumably via cytoreduc-
lung cancer results in an overall5-year survival tion, destruction of adjacent sensory neural
rate of 34% in the geriatric population (50). fibers, and decreased neural stimulation sec-
Radiofrequency ablation may be applied to ondary to debulking (27,29,55-57). The resul-
these patients and to certain patients in whom tant improved quality of life illustrates the
a small number of slowly growing metastases dynamic use of this procedure, but more con-
are identified. In particular, metastases from trolled studies need to be done. Callstrom et al
renal cell carcinoma, sarcoma, and colorectal (57) have reported palliative effects within
carcinoma are those that will benefit from this 1 week of RFA of osseous metastases, with
therapy (46,47). One-third of patients with increasing benefit and quality of life directly
renal cell carcinoma have pulmonary metas- related to time.
tases [60% with metastasis in general (45)], and
one-half of patients who have undergone
nephrectomy for renal cell carcinoma will Conclusion
develop pulmonary metastases later (15). Data
from several groups presented in abstract form The minimally invasive technique of RFA has
(51) have shown early success with RFA for a promising impact in the treatment of nonsur-
colorectal metastases. Tumor control has been gical patients of lung tumors, provided that
achieved in approximately 90% in RFA of colo- control of local disease will improve quality of
rectal hepatic metastases (52). However, long- life, survival, and overall prognosis. It may
term outcome data are not available. become a procedure for curative as well as pal-
The exact size and number of lesions have yet liative intent, either solely or as an adjunct to
to be defined, but it is not unreasonable to use current standard modalities. Advantages of
similar parameters that have been applied to RFA include precise control, low cost, accept-
liver tumors (i.e., four or fewer metastases). The able morbidity and mortality as well as compli-
maximum size for effective treatment has not cation rate, and use in the outpatient setting.
been established, but again, lesions less than The potential paradigm shift in standard treat-
3 cm in size (as in the liver) are optimal candi- ment toward use of alternative methods such as
dates, given the treatment regions achievable RFA depends on precise scientific and clinical
with current RFA technology (Fig. 28.5). His- research/data to provide evidence-based
tologic evidence of complete lethal thermal medicine. Long-term follow-up elucidating the
injury in pulmonary metastases by RFA has mechanism and effect of this procedure await
been shown (53,54). As with most cancers, early further studies.
28. Radiofrequency Ablation for Thoracic Neoplasms 363

E F
FIGURE 28.5. A 68-year-old man with metastatic Both resolved without any intervention. (D) Axial
renal cell carcinoma to the left kidney, brain, and CT image showing a 1.7-cm spiculated nodule in the
lungs, status post-wedge resection. (A) Axial CT right lower lobe (arrow). (E) RF electrode (arrow)
image showing a pulmonary nodule in the right within the tumor bed. (F) Axial CT image immedi-
upper lobe (arrow). (B) RFA of the tumor with the ately postprocedure revealed a small pneumothorax
electrode (arrow) in the nodule. (C) Axial CT image as well as hemorrhage and air space opacity (arrow)
immediately postprocedure revealed a small pneu- around the nodule. Both resolved without any
mothorax (small arrow) as well as hemorrhage and intervention.
air space opacity (larger arrow) around the nodule.
364 S.K. Jain and nE. Dupuy

Ga Gb

Gc Gd
FIGURE 28.5. (G) Serial axial CT images at 3-year with stable subcentimeter nodules (small arrows) in
follow-up showing stable RFA site of lung metas- the right lung.
tases in the right upper and lower lobes (large arrow)
28. Radiofrequency Ablation for Thoracic Neoplasms 365

A 8

FIGURE 28.6. A 40-year-old man with metastatic lung the tumor. Arrow indicates the electrode within
cancer status post-left pneumonectomy with adju- the lesion. (C,D) Follow-up magnetic resonance
vant chemotherapy and radiation referred for RFA imaging. TI-weighted gradient echo postcontrast
for chest wall palliation. (A) A 4.0 x 2.0 x 2.2cm images at I-month (C) and 3-month (D) follow-up.
expansile, mixed lytic and sclerotic rib metastasis in The latter (arrow) shows reduced enhancement in
the right anterolateral rib cage (arrow). (B) RFA of comparison to the former.
366 S.K. Jain and D.E. Dupuy

References 16. Kandioler D, Kromer E, Tuchler H, et al. Long-


term results after repeated surgical removal of
1. Cancer Facts and Figures 2003. Atlanta: Ameri- pulmonary metastases. Ann Thorac Surg 1998;
can Cancer Society, 2003. 65:909-912.
2. Mountain CF. Revisions in the International 17. Ginsberg RJ, Rubinstein LV. Randomized trial of
System for Staging Lung Cancer. Chest 1997; lobectomy versus limited resection for T1 NO
111:1710-1717. non-small cell lung cancer. Lung Cancer Study
3. Van Rens M, de la Riviere AB, Elbers HRJ, et Group. Ann Thorac Surg 1995;60:615-623.
al. Prognostic assessment of 2,361 patients who 18. Kvale RA, Simoff M, Prakash UBS. Palliative
underwent pulmonary resection for non-small care. Chest 2003;123:284S-311S.
cell lung cancer, stage I, II, and IlIA. Chest 2000; 19. Griffin JP, Nelson JE, Koch KA, et al. End-of-life
117:374-379. care in patients with lung cancer. Chest 2003;123:
4. Rajdev L, Keller SM. Neoadjuvant and adjuvant 312S-331S.
therapy of non-small cell lung cancer. Surg 20. SUPPORT principal investigators. A controlled
Oncol 2002;11:243-253. trial to improve care for seriously ill hospitalized
5. Dupuy DE, Zagoria RJ, Akerley W, Mayo-Smith patients. JAMA 1995;274:1591-1598.
W, Kavanagh PV, Safran H. Percutaneous 21. Downey RJ, Martini N, Rusch V, et al. Extent of
radiofrequency ablation of malignancies in the chest wall invasion and survival in patients with
lung. AJR 2000;174:57-59. lung cancer. Ann Thorac Surg 1999;68:188-193.
6. Goldberg SN, Dupuy DE. Image-guided 22. Watson PN, Evans RJ. Intractable pain with lung
radiofrequency tumor ablation: challenges and cancer. Pain 1987;29:163-173.
opportunities-part I. J Vasc Intervent Radiol 23. Jacox A, Carr DB, Payne R, et al. Management
2001;12:1021-1032. of cancer pain: clinical practice guideline No.9.
7. Dupuy DE, Goldberg SN. Image-guided Rockville, MD: Agency for Health Care Policy
radiofrequency tumor ablation: challenges and and Research, U.S. Department of Health and
opportunities-part II. J Vasc Intervent Radiol Human Services, Public Health Service, March
2001;12:1135-1148. 1994; AHCPR Publication No. 94-0592.
8. Murai N, Koga K, Nagamachi S, et al. Radio- 24. Langendijk JA, Ten Velde GPM, Aaronson NK,
therapy in bone metastases, with special refer- et al. Quality of life after palliative radiotherapy
ence to its effect on relieving pain. Gan No in non-small cell lung cancer: a prospective
Rinsho 1989;35:1149-1152. study. Int J Radiat Oncol BioI Phys 2000;47:
9. Highland AM, Mack P, Breen DJ. Radiofre- 149-155.
quency thermal ablation of a metastatic lung 25. Dupuy DE, Mayo-Smith WW, Abbott GF,
nodule. Eur Radiol 2002;suppl4:S166-S170. DiPetrillo T. Clinical applications of radio-
10. Hoegler D. Radiotherapy for palliation symp- frequency tumor ablation in the thorax. Radio-
toms in incurable cancer. Curr Probl Cancer Graphies 2002;22:S259-S269.
1997;21:129-183. 26. Warzelhan J, Stoelben E, Imdahl A, et al. Results
11. Todd TR. The surgical treatment of pulmonary in surgery for primary and metastatic chest wall
metastases. Chest 1997;112:287S-290S. tumors. Eur J Cardiothorac Surg 2001;19:584-
12. Pastorino U, Buyse M, Friedel G, et al. Long- 588.
term results of lung metastasectomy: prognostic 27. Patti J, Neeman Z, Wood B. Radiofrequency
analyses based on 5206 cases. J Thorac Cardio- ablation for cancer-associated pain. J Pain 2002;
vasc Surg 1997;113:37--49. 3:471.
13. Sakamoto T, Tsubota N,Ivanaga K, et al. Pul- 28. McCaughan B. Recent advances in managing
monary resection for metastases from colorectal non-small-cell lung cancer: 2. Med J Aust 1997;
cancer. Chest 2001;119:1069-1072. 166(suppl):S7-SlO.
14. Fourquier P, Regnard J, Rea S, et al. Lung metas- 29. Dupuy DE, Safran H, Mayo-Smith WW,
tases of renal cell carcinoma: results of surgical Goldberg SN. Radiofrequency ablation of
resection. Eur J Cardiothorac Surg 1997;11: painful osseous metastatic disease: scientific
17-21. paper presented at RSNA annual meeting
15. Dekernion JB, Ramming KP, Smith RB. The Chicago. Radiology 1998;209S:171S-172S.
natural history of metastatic renal cell carci- 30. Kapural L, Mekhail N. Radiofrequency ablation
noma: a computer analysis. J Urol 1978;120: for chronic pain control. Current Pain Headache
148-152. Rep 2001;5:517-525.
28. Radiofrequency Ablation for Thoracic Neoplasms 367

31. Elia S, Griffo S, Gentile M, et a!. Surgical treat- 45. van der Poel HG, Roukema JA, Horenblas S,
ment of lung cancer invading chest wall: a van Geel AN, Debruyne FM. Metastasectomy in
retrospective analysis of 110 patients. Eur J renal cell carcinoma: a multicenter retrospective
Cardiothorac Surg 2001;20:356-360. analysis. Eur Urol 1999;35:197-203.
32. Sawada M, Watanabe S, Tsuda H, et a1. An 46. Piltz S, Meimarakis G, Wichmann MW, et a1.
increase in body temperature during radiofre- Long-term results after pulmonary resection
quency ablation of liver tumors. Anesth Analg of renal cell carcinoma metastases. Ann Thorac
2002;94:1416-1420. Surg 2002;73:1082-1087.
33. Landreneau RJ, Sugarbaker DJ, Mack MJ, et a1. 47. Okumura S, Kondo H, Tsuboi M, et a1. Pul-
Wedge resection versus lobectomy for stage monary resection for metastatic colorectal
1(T! NO MO) non-small cell lung cancer. cancer: experience with 159 patients. J Thorac
J Thorac Cardiovasc Surg 1997;113:691-698. Cardiovasc Surg 1996;112:867-874.
34. Martini N, Bains MS, Burt ME, et a1. Incidence 48. Labow DM, Buell JE, Yoshida A, Rosen S,
of local recurrence and second primaries in Posner Me. Isolated pulmonary recurrence after
resected stage I lung cancer. J Thorac Cardiovasc resection of colorectal hepatic metastases-is
Surg 1995;109:120-129. resection indicated? Cancer J 2002;8:342-347.
35. Mountain CE A new international staging 49. Hamy A, Baron 0, Bennouna J, Roussel JC,
system for lung cancer. Chest 1986;89:225- Paineau J, Douillard JY. Resection of hepatic
233. and pulmonary metastases in patients with colo-
36. Naruke T, Goya T, Tsuchiya R, et a1. Prognosis rectal cancer. Am J Clin Oncol 2001;24:607-
and survival in resected lung carcinoma based on 609.
the new international staging system. J Thorac 50. Gauden S, Tripcony L. The curative treatment by
Cardiovasc Surg 1988;96:440-447. radiation therapy alone of stage I non-small cell
37. Shimizu J, Watanabe Y, Oda M, et a1. Results of lung cancer in a geriatric population. Lung
surgical treatment of stage I lung cancer. Nippon Cancer 2001;32:71-79.
Geka Gakkai Zasshi 1993;94:505-510. 51. Glenn DW, Clark W, Morris DL, et a1. Percuta-
38. Williams DE, Pairolero PC, Danis CS, et a1. neous radiofrequency ablation of colorectal pul-
Survival of patients surgically treated for stage monary metastases. In: RSNA Conference 2001,
I lung cancer. J Thorac Cardiovasc Surg 1981; November 25-30, Chicago (Abstract 391).
82:70-76. 52. de Baere T, Elias D, Dromain C, et a1. Radiofre-
39. Martini N. Surgical treatment of non-small cell quency ablation of 100 hepatic metastases with
lung cancer by stage. Semin Surg Oncol 1990;6: a mean follow-up of more than 1 year. AJR
248-254. 2001;175:1619-1625.
40. Kodama K, Doi 0, Higashiymama M, et a!. 53. Steinke K, Habicht JM, Thomsen S, Soler M,
Intentional limited resection for selected Jacob AL. CT-guided radiofrequency ablation of
patients with T1 NO non-small cell lung cancer: a pulmonary metastasis followed by surgical
a single-institution study. J Thorac Cardiovasc resection. Cardiovasc Intervent Radiol 2002;25:
Surg 1997;114:347-353. 543-546.
41. Okada M, Yoshikawa K, Hatta T, et a1. Is seg- 54. Miao Y, Yicheng N, Bosmans H, et a1. Radiofre-
mentectomy with lymph node assessment an quency ablation for eradication of pulmonary
alternative to lobectomy for non-small cell lung tumor in rabbits. J Surg Res 2001;99:265-271.
cancer of 2 cm or smaller? Ann Thorac Surg 55. Kishi K, Nakamura H, Sudo A, et a1. Tumor
2001;71:956-960. debulking by radiofrequency ablation in hyper-
42. Wright SE. Adjuvant chemotherapy and radia- trophic pulmonary osteoarthropathy associated
tion therapy for elimination of residual micro- with pulmonary carcinoma. Lung Cancer 2002;
scopic non-small cell lung cancer. Int J Oncol 38:317-320.
1999;14:347-351. 56. Ohhigashi S, Nishio T, Watanabe F, et a1. Experi-
43. Okamoto Y, Murakami M, Yoden E, et a1. Reir- ence with radiofrequency ablation in the treat-
radiation for locally recurrent lung cancer previ- ment of pelvic recurrence in rectal cancer: report
ously treated with radiation therapy. Int J Radiat of two cases. Dis Colon Recum 2001;44:741-745.
Oncol BioI Phys 2002;52:390-396. 57. Callstrom MR, Charboneau JW, Goetz Mp, et a1.
44. Temple LK, Brennan ME The role of pulmonary Painful metastases involving bone: feasibility
metastasectomy in soft tissue sarcoma. Semin of percutaneous CT- and US-guided radio-
Thorac Cardiovasc Surg 2002;14:35-44. frequency ablation. Radiology 2002;224:87-97.
368 S.K. Jain and D.E. Dupuy

58. Anderson BO, Burt ME. Chest wall neoplasms cancer from 1976 through 1990. J Thorac
and their management. Ann Thorac Surg 1994; Cardiovasc Surg 1996;112:349-355.
58:1774-1781. 61. Flehinger BJ, Kimmel M, Melamed MR. The
59. Pearson FG. Lung cancer: the past twenty-five effect of surgical treatment on survival from
years. Chest 1986;89 (4 suppl):200S-205S. early lung cancer: implications for screening.
60. Wada H, Tanaka F, Yanagihara K. Time trends Chest 1992;101:1013-1018.
and survival after operations for primary lung
29
Soft Tissue Ablation
Sridhar Shankar, Eric vanSonnenberg, Stuart G. Silverman, Paul R. Morrison,
and Kemal Tuncali

Overview mas have been successfully treated percuta-


neously with CT-guided RFA for several years
Over the past several years, percutaneous (14-16). More recently, ultrasound and CT-
image-guided tumor ablation procedures have guided RFA has been suggested as a method
proliferated (1-7). Several new technologies, in for the palliation of painful metastases involv-
addition to the previously available percuta- ing bone and soft tissues (7,17-20).
neous injection of toxic substances (such as
alcohol, hot saline, acetic acid), have been
added to the interventional radiologist's
armamentarium. These technologies include Definitions
radiofrequency ablation (RFA) , laser (laser
interstitial tumor therapy, UTI), cryotherapy, Ablation of soft tissue tumors is not new;
microwave therapy, and high-intensity focused animal studies using tumor models (e.g., VX2
ultrasound (HIFU) therapy; all except HIFU tumor model, rat breast cancer) use ablation of
are percutaneous techniques, and HIFU is trans- artificially induced soft tissue tumors to assess
cutaneous (8). Several centers are now using efficacy of various ablative techniques (21-24).
these techniques clinically, and a large body of Soft tissue lesions comprise those that are
experience has accumulated. While most of situated in no particular organ system; the
these ablative techniques have been used pre- closest, perhaps, is the musculoskeletal system,
dominantly for the treatment of malignant liver although many lesions lie outside that system
tumors, new sites and indications continue to per se. Several lesions are inseparable from the
emerge. underlying, or closely related bone; indeed, sit-
The impetus for treating tumors in extrahe- uations are common in which the lesion is part
patic sites is based mostly on clinical trials that soft tissue and part bone. Many of these lesions
have demonstrated percutaneous ultrasound can more properly be considered under the
(US) and computed tomography (CT) guid- umbrella term soft tissue and bone lesions.
ance, predominantly using RFA to be safe and Clinically the most important situation would
effective in selected patients with liver lesions likely be a malignant lesion, and examples
(4,9). Consequently, RFA is now being used include recurrent tumors in the resection bed
as an alternative to conventional treatment of intraabdominal (including retroperitoneal
(i.e., surgery, radiation, and chemotherapy) for tumors), pelvic, thoracic, extremity, and head
tumors in several other organ systems, includ- and neck cancers. Some benign lesions such as
ing lung, kidney, bone, and soft tissue, in certain arteriovenous malformations (11) also may be
select situations (10-13). Indeed, osteoid osteo- included in this category.

369
370 S. Shankar et al

Indications ablations in the head and neck region are avail-


able. Most of these detail the performance of
Most patients who are referred for percuta- ablation on a case-by-case basis (25,26).
neous ablation of soft tissue tumors are candi- Alcohol injection has been successfully and
dates for palliation only. Commonly, referrals safely performed in metastatic thyroid cancer
are for local tumor and/or pain control, and few to cervical lymph nodes (27). We recently pal-
patients realistically have the possibility for liatively injected alcohol into large cervical
cure. Less common indications include inter- nodes in a patient with metastatic small-cell
ference with function of a limb or organ, and lung cancer for pain relief with successful short-
paraneoplastic symptoms that are believed to term results (Fig. 29.1). Radiofrequency abla-
be secondary to humoral factors elaborated by tion of recurrent thyroid cancer also has been
the tumor (The referral in this case would be to reported (28). The factor of overriding impor-
debulk the tumor to control the paraneoplastic tance in these situations appears to be the care
syndrome.). Most patients already have had taken to avoid or minimize damage to adjacent
surgical resection, with or without palliative critical structures, such as nerves and vessels
radiotherapy, and many patients are either that are present in the head and neck region.
undergoing chemotherapy presently or have Intermittent intraprocedural neurologic exam-
done so previously. ination during conscious sedation is one way to
be forewarned of this possibility (29). Also, care
must be taken to avoid damage to the skin, as
many soft tissue tumors are located superfi-
Sites of Application and cially (30,31).
Potential Complications
Moving sequentially from the head to the foot, Thorax
soft tissue ablation has myriad current and There are several published studies on ablation
potential applications: of lung and thoracic tumors; several of the
ablated tumors extend beyond the confines of
the lung, and invade the pleura, ribs, and adja-
Head and Neck
cent soft tissues. Many of these procedures are
Several case reports and small series that detail performed for palliation of pain, and could be
percutaneous, intraoperative, and laparoscopic included under the purview of soft tissue tumor

A B
FIGURE 29.1. A 70-year-old woman with metastatic (B) Immediate postprocedure contrast-enhanced
small-cell lung cancer and bulky cervical lymph- computed tomography (CT) scan following injection
adenopathy. (A) A 22-gauge needle (arrowheads) of 20cc of alcohol demonstrates low-density central
within the left-sided supraclavicular nodal mass. necrosis (arrows).
29. Soft Tissue Ablation 371

ablation (32,33). Pain control usually is


achieved by ablating the portion of the tumor
that invades the chest wall; we have found
adjunctive intercostal nerve block to be useful
for pain control as well (32).

Abdomen
Retroperitoneal tumors comprise the common-
est extravisceral site in this category. Repre-
sentative tumors include leiomyosarcoma,
usually recurrent following primary resection, A
transitional cell cancer, renal cell cancer, colon
cancer, and metastatic and nodal disease from
a variety of primaries (13,20). We have per-
formed both alcohol injection and RFA
(Fig. 29.2) in the retroperitoneum. Close to the
pancreas, care should be taken not to cause
alcohol leak or thermal injury that might result
in pancreatitis.

Pelvis
A common situation for ablation is presacral B
recurrence of rectal cancer following abdom-
inoperineal resection. It is a good practice to FIGURE 29.2. A 73-year-old woman with metastatic
gastric cancer with a conglomerate lymph node
biopsy the lesion at a separate session prior to mass in the retroperitoneum. (A) CT image during
performing the ablation, even in the presence RFA; probe is seen positioned within the retroperi-
of fairly convincing imaging evidence of tumor toneal nodal mass (arrows). Small specks of gas
recurrence. In our experience, some of these are seen surrounding the probe, a common finding.
lesions have been shown to be of infectious, (B) Postprocedure contrast-enhanced MR image
postsurgical scarring, or some other benign demonstrates a predominantly nonenhancing mass.
etiology, proven by percutaneous biopsy and Central low-intensity area is a susceptibility artifact.
imaging follow-up (Fig. 29.3).
Caution should be exercised when ablating
lesions located in the inguinal region, as in tases. Metastatic tumors from primary tumors
many instances these lesions involve nerves such as lung and breast cancer to bone and soft
that cannot be visualized using current imaging tissues can be painful; percutaneous ablation
techniques. We have encountered an injury to of these lesions can be a worthwhile palliative
the femoral nerve from ablation of a soft tissue treatment for these patients (17) (Figs. 29.4 and
sarcoma in the groin with RF. Indeed, it is 29.5). We have performed palliative magnetic
essential that patients be informed about the resonance imaging (MRI) guided cryoablation
potential for nerve injury and limb paralysis in a patient with metastatic mesothelioma to
during the consent process. both the humerus and femur with good pain
control achieved. Other sites include lesions
involving, or close to, the vertebral column that
Extremities and Other Sites are either painful or cause pressure symptoms
Many primary tumors of the extremities recur on adjacent nerves (11,34).
after local resection, and some of these patients Menendez et al (35) reported 12 cases
are not candidates for re-resection for medical of extremity sarcomas that they treated with
reasons or due to the presence of other metas- intraoperative US-guided cryotherapy, and
372 S. Shankar et al

c
A
FIGURE 29.3. A 50-year-old woman with locally recurrent rectal
cancer in the presacral area. She had undergone abdominoper-
ineal resection of the primary tumor 6 years earlier. (A) MR-
guided cryoablation of the presacral tumor-the iceball is seen as
an exquisitely well-defined area of signal void in this prone axial
scan (arrows). Note cryoprobe (curved arrow) entering from the
right gluteal region. (B) Contrast-enhanced MR image of the
pelvis obtained approximately 12 months following the proce-
dure demonstrates irregular enhancing areas in the presacral
region (arrows). The previously ablated area has decreased in
size. (C) CT-guided fan biopsy of suspicious areas in the presacral
region. Pathology revealed fibroconnective tissue with necrosis,
acute inflammation, and granulation tissue, but no evidence of
B tumor.

A B
FIGURE 29.4. A 45-year-old man with locally recur- evidence of local recurrence. The patient sustained
rent, painful sarcoma in the left groin. (A) CT-guided injury to the femoral nerve with paresis of his quadri-
RFA of the left groin tumor. (B) Eight-month ceps; he has slowly recovered with the aid of physi-
follow-up contrast-enhanced MR exam demon- cal therapy.
strates no enhancement in the treated tumor, and no
29. Soft Tissue Ablation 373

urethra while treating lesions that invade the


prostate (Fig. 29.6), bladder base, or are other-
wise situated too close to the urethra; this is
standard technique for prostate cryotherapy
(36).
Percutaneously introduced saline, carbon
dioxide (37,38), and possibly balloons or other
mechanical devices may be helpful in moving
structures such as the colon, kidney, or spleen
out of the area to be treated, such that both
complete ablation may be achieved and injury
A to bowel avoided.
Injury to overlying skin, if too close to the
ablative site, may be prevented by using heated
or cooled saline moistened gauze packs to
prevent thermal injury. We use warm saline-
soaked gauze packs routinely to keep the skin
from freezing while treating lesions close to
the surface with cryotherapy. Conversely, cool
packs may also be useful for lowering the skin
temperature during RFA of lesions close to the
surface.
It may be worthwhile to remember that
B larger volumes of tumor necrosis with soft
FIGURE 29.5. A 48-year-old woman with metastatic
leiomyosarcoma to the left gluteal region. (A) CT-
guided RFA of the left gluteal mass (arrows).
(B) Postprocedure contrast-enhanced CT scan
demonstrates the ablated area (arrows) to be
nonenhancing.

concluded that the technique was safe and fea-


sible. They encountered three patients with
nerve palsy, all of whom recovered completely.

Special Situations and


Adjunctive Procedures
for Ablation
Tumors located adjacent to critical structures
demand measures beyond customary caution to
treat the lesion successfully. Thus, preproce-
FIGURE 29.6. Locally recurrent rectal cancer invad-
dural ureteral stenting can help avoid damage
ing the prostate gland. MR-guided cryotherapy with
to the ureter while treating lesions in proximity a urethral warming catheter within the urethra
to it. The urologist performs the ureteral stent- (prone oblique image). Note indentation of the
ing cystoscopically, one day prior to or on iceball adjacent to the urethral warming catheter
the day of the procedure. Urethral warming (arrow). The ischial tuberosities are seen on either
catheters can be used to prevent injury to the side of the iceball (open arrows).
374 S. Shankar et al

tissue lesions than tumors in the liver some- week intervals to evaluate efficacy of treatment
times may be achieved with RFA. This is and tumor recurrence. The rationale for using
because most standard RF algorithms are multiple modalities for lesion assessment is that
created for the liver, which is liberally perfused. some tumors are poorly vascularized, and many
The larger lesion sizes achieved in soft tissue lesions demonstrate irregular areas of intratu-
lesions may be secondary to these tumors being moral necrosis that may add to confusion while
less well vascularized, differences in water assessing the postprocedure thermal effect.
content, or electrical and thermal conductivity Pain evaluation can be performed using the
of the tumor tissue (29,39). Wisconsin Brief Pain Questionnaire, which
Larger volumes of tissue necrosis also may rates the patient's pain on a scale of 0 to 10,
be achieved by percutaneous injection of with 0 being no pain and 10 the worst pain the
alcohol into the tumor prior to performing patient has ever experienced (43,44).
RFA (22). We have found this technique useful
in obtaining significantly larger volumes of
tumor necrosis in the liver (40). Conclusions
Many malignant tumors of soft tissues can be
treated percutaneously using a variety of tech-
Techniques, Guidance niques and devices. These therapies may
Modalities, and Follow-Up provide a means for the palliation of pain unre-
sponsive to other therapies, in addition to local
In general, the techniques and guidance control of tumor. Care with structures either
systems that are utilized elsewhere in the body within (nerves, vessels) or juxtaposed to
can be directly extrapolated to soft tissue (bowel, urethra, ureter) these soft tissue tumors
tumors. As discussed above, different body must be exercised, as these are distinct hazards
parts have different requirements in terms of in the treatment of these lesions.
precautions and ancillary measures. Computed
tomography and US are the most commonly References
utilized modalities to target tumors and guide
needle/probe placement. The major disadvan- 1. Livraghi T, Goldberg SN, Lazzaroni S, et al.
tage with both imaging techniques is that they Hepatocellular carcinoma: radio-frequency abla-
do not allow monitoring of the effects of RFA tion of medium and large lesions. Radiology
2000;214:761-768.
in sufficient detail to allow the operator to visu-
2. Livraghi T, Goldberg SN, Solbiati L, Meloni F,
alize the margins of the ablation. As a result, Ierace T, Gazelle GS. Percutaneous radio-
percutaneous US- or CT-guided RFA may be frequency ablation of liver metastases from
hazardous in patients with metastatic or re- breast cancer: initial experience in 24 patients.
current tumors of soft tissue or bone situated Radiology 2001;220:145-149.
adjacent to critical structures such as bowel, 3. Curley SA, Izzo F. Radiofrequency ablation of
bladder, and nerves. Magnetic resonance primary and metastic liver tumors. Surg Technol
imaging (MRI)-guided percutaneous cryother- Int 2002;10:99-106.
apy may be the most elegant technique avail- 4. Gazelle GS, Goldberg SN, Solbiati L, Livraghi T.
able in terms of visualization of tumor, Tumor ablation with radio-frequency energy.
guidance, treatment monitoring, and follow-up Radiology 2000;217:633-646.
5. Gervais DA, McGovern FJ, Wood BJ, Goldberg
currently (41,42).
SN, McDougal WS, Mueller PRo Radio-
Follow-up of treated soft tissue tumors is per- frequency ablation of renal cell carcinoma: early
formed by a combination of various imaging clinical experience. Radiology 2000;217:665-672.
modalities. We utilize CT and MRI in the imme- 6. Solbiati L, Goldberg SN, Ierace T, et al. Hepatic
diate postprocedure period, and CT, MRI, and metastases: percutaneous radio-frequency abla-
positron emission tomography (PET) at base- tion with cooled-tip electrodes. Radiology 1997;
line (preprocedure), and at approximately 12- 205:367-373.
29. Soft Tissue Ablation 375

7. Wood BJ, Fojo A, Levy EB, Gomez-Horhez J, cutaneous radiofrequency ablation of spinal
Chang R, Spies 1. Radiofrequency ablation of tumors. European Congress of Radiology, 2001.
painful neoplasms as a palliative therapy: early 20. Shankar S, Tuncali K, vanSonnenberg E,
experience.2000;11(S):207. Morrison PR, Kacher D, Silverman So. MRI
8. Hynynen K, Pomeroy 0, Smith DN, et al. MR guided percutaneous cryotherapy of soft tissue
imaging-guided focused ultrasound surgery of and bone metastases. Radiology 2001;221(P):
fibroadenomas in the breast: a feasibility study. 558.
Radiology 2001;219:176-185. 21. Hazle JD, Stafford RJ, Price RE. Magnetic
9. Goldberg SN, Gazelle GS, Mueller PRo Thermal resonance imaging-guided focused ultrasound
ablation therapy for focal malignancy: a unified thermal therapy in experimental animal models:
approach to underlying principles, techniques, correlation of ablation volumes with pathology
and diagnostic imaging guidance. AJR 2000;174: in rabbit muscle and VX2 tumors. J Magn Reson
323-331. Imaging 2002;15:185-194.
10. Wood BJ, Ramkaransingh JR, Fojo T, Walther 22. Goldberg SN, Kruskal JB, Oliver BS, Clouse ME,
MM, Libutti SK. Percutaneous tumor abla- Gazelle GS. Percutaneous tumor ablation:
tion with radiofrequency. Cancer 2002;94:443- increased coagulation by combining radio-
451. frequency ablation and ethanol instillation in a
11. vanSonnenberg E, Hadjipavlou A, Chaljub G, rat breast tumor model. Radiology 2000;217:
Nolsoe C, Ko E. Therapeutic cryotherapy guided 827-831.
by MRI and ultrasound for vascular malforma- 23. Monsky WL, Kruskal JB, Lukyanov AN, et al.
tions in erector muscles of the back. Min Invas Radio-frequency ablation increases intratumoral
Ther Allied TechnoI1997;6:343-348. liposomal doxorubicin accumulation in a rat
12. Nader R, Alford BT, Nauta HJ, Crow W, van- breast tumor model. Radiology 2002;224:823-
Sonnenberg E, Hadjipavlou Ao. Preoperative 829.
embolization and intraoperative cryocoagula- 24. Ahmed M, Lobo SM, Weinstein J, et al.
tion as adjuncts in resection of hypervascular Improved coagulation with saline solution pre-
lesions of the thoracolumbar spine. J Neurosurg treatment during radiofrequency tumor ablation
2002;97:294-300. in a canine model. J Vasc Intervent Radiol 2002;
13. Gervais DA, Arellano RS, Mueller PRo Percuta- 13:717-724.
neous radiofrequency ablation of nodal metas- 25. Bui QT, Dupuy DE. Percutaneous CT-guided
tases. Cardiovasc Intervent Radiol 2002;25: radiofrequency ablation of an adenoid cystic car-
547-549. cinoma of the head and neck. AJR 2002;179:
14. Rosenthal DL Percutaneous radiofrequency 1333-1335.
treatment of osteoid osteomas. Semin Muscu- 26. Owen RP, Ravikumar TS, Silver CE, Beitler J,
loskel Radiol 1997;1:265-272. Wadler S, Bello 1. Radiofrequency ablation of
15. Rosenthal DI, Alexander A, Rosenberg AE, head and neck tumors: dramatic results from
Springfield D. Ablation of osteoid osteomas with application of a new technology. Head Neck
a percutaneously placed electrode: a new proce- 2002;24:754-758.
dure. Radiology 1992;183:29-33. 27. Lewis BD, Hay ID, Charboneau JW, Mciver B,
16. Rosenthal DI, Hornicek FJ, Wolfe MW, Jennings Reading CC, Goellner JR. Percutaneous ethanol
LC, Gebhardt MC, Mankin H1. Percutaneous injection for treatment of cervical lymph node
radiofrequency coagulation of osteoid osteoma metastases in patients with papillary thyroid
compared with operative treatment. J Bone Joint carcinoma. AJR 2002;178:699-704.
Surg [Am] 1998;80:815-821. 28. Dupuy DE, Monchik JM, Decrea C, Pisharodi L.
17. Callstrom MR, Charboneau JW, Goetz MP, et al. Radiofrequency ablation of regional recurrence
Painful metastases involving bone: feasibility from well-differentiated thyroid malignancy.
of percutaneous CT- and US-guided radio- Surgery 2001;130:971-977.
frequency ablation. Radiology 2002;224:87-97. 29. Neeman Z, Patti JW, Wood B1. Percutaneous
18. Dupuy DE, Safran H, Mayo-Smith WW, radiofrequency ablation of chordoma.AJR 2002;
Goldberg SN. Radiofrequency ablation of 179:1330-1332.
painful osseous metastatic disease. Radiology 30. Kanpolat Y, Savas A, Bekar A, Berk C. Per-
1998;209(P):389. cutaneous controlled radiofrequency trigeminal
19. Gevargez A, Matysek M, Kriener PG, Siepmann rhizotomy for the treatment of idiopathic
G, Braun M, Gronemeyer DHW. CT guided per- trigeminal neuralgia: 25-year experience with
376 S. Shankar et al

1600 patients. Neurosurgery 2001;48:524-532; 38. Langen HJ, Jochims M, Gunther RW. Artificial
discussion 532-534. displacement of kidneys, spleen, and colon by
31. Onofrio BM. Radiofrequency of percutaneous injection of physiologic saline and CO2 as an aid
Gasserian ganglion lesions. Results in 140 pa- to percutaneous procedures: experimental
tients with trigeminal pain. J Neurosurg 1975; results. J Vase Intervent Radiol1995;6:411-416.
42:132-139. 39. Dupuy DE, Goldberg SN. Image-guided
32. vanSonnenberg E, Shankar S, Tuncali K, radiofrequency tumor ablation: challenges and
Silverman SG, Morrison PR, Jaklitsch M. Initial opportunities-part II. J Vase Intervent Radiol
clinical experience with RF ablation of lung 2001 ;12:1135-1148.
tumors. Radiology 2002;225(P):291. 40. Shankar S, vanSonnenberg E, Silverman SG,
33. Dupuy DE, Mayo-Smith WW, Abbott GF, Tuncali K, Morrison PRo Combined radiofre-
DiPetrillo T. Clinical applications of radio- quency (RF) and direct alcohol infusion for per-
frequency tumor ablation in the thorax. cutaneous tumor ablation. AJR 2004;183:1425-
Radiographies 2002;22:S259-269. 1429.
34. Dupuy DE, Hong R, Oliver B, Goldberg SN. 41. Shankar S, vanSonnenberg E, Silverman SG,
Radiofrequency ablation of spinal tumors: tem- Tuncali K. Interventional radiology procedures
perature distribution in the spinal canal. AJR in the liver. Biopsy, drainage, and ablation. Clin
2000;175:1263-1266. Liver Dis 2002;6:91-118.
35. Menendez LR, Tan MS, Kiyabu MT, Chawla SP. 42. Silverman SG, Tuncali K, Adams DF, et al. MR
Cryosurgical ablation of soft tissue sarcomas: a imaging-guided percutaneous cryotherapy of
phase I trial of feasibility and safety. Cancer liver tumors: initial experience. Radiology 2000;
1999;86:50-57. 217:657-664.
36. Weider J, Schmidt JD, Casola G, vanSonnenberg 43. Daut RL, Cleeland CS, Flanery RC. Develop-
E, Stainken BF, Parsons CL. Transrectal ment of the Wisconsin Brief Pain Questionnaire
ultrasound-guided transperineal cryoablation in to assess pain in cancer and other diseases. Pain
the treatment of prostate carcinoma: preliminary 1983;17:197-210.
results. J Urol 1995;154:435-441. 44. Daut RL, Cleeland CS. The prevalence and
37. Goodacre BW, Savage C, Zwischenberger JB, severity of pain in cancer. Cancer 1982;50:
Wittich GR, vanSonnenberg E. Salinoma 1913-1918.
window technique for mediastinal lymph node
biopsy. Ann Thorac Surg 2002;74:276-277.
30
Image-Guided Palliation of Painful
Skeletal Metastases
Matthew R. Callstrom, 1. William Charboneau, Matthew P. Goetz,
and Joseph Rubin

Painful skeletal metastases are a common patients; however, 20% to 30% of patients
problem in patients with cancer. Autopsy studies treated with this modality do not experience
have shown up to 85% of patients who die pain relief, and few options exist for these
from breast, prostate, and lung cancer have evi- patients (12-17). Furthermore, patients who
dence of bone metastases at the time of death have recurrent pain at a previously irradiated
(1). Skeletal metastases often result in com- metastatic site may not be eligible for further
plications such as pain, fractures, and decreased RT because of limitations in normal tissue tol-
mobility that adversely affect a patient's erance. Unfortunately, skeletal disease in this
quality of life, and ultimately reduce per- patient population often is refractory to stan-
formance status. In addition, these complica- dard chemotherapy or hormonal therapy.
tions can affect a patient's mood and lead Surgery, which is usually reserved for impend-
to associated depression and anxiety (1, 2). ing fracture, is not always an option when
Current treatment for patients with bone metas- patients present with advanced disease and
tases is primarily palliative and includes the poor functional status. Radiopharmaceuticals,
following: localized therapies (radiation and which have known benefit in patients with
surgery), systemic therapies (chemotherapy, diffuse painful bony metastases, are not consid-
hormonal therapy, radiopharmaceuticals, and ered the standard of care for patients with iso-
bisphosphonates), and analgesics (opioids and lated, painful lesions. For many patients with
nonsteroidal antiinflammatory drugs). painful metastatic disease, analgesics remain
The causes of pain in patients with bone the only alternative treatment option. Unfortu-
metastases are not fully understood, and the nately, to obtain sufficient pain control for many
presence of pain is not correlated with the of these patients, side effects such as constipa-
type of tumor, location, number, or size of the tion, nausea, and sedation can be significant.
metastases (2-4). Possible mechanisms of pain Many patients with pain from metastatic
include: (1) stretching of the periosteum sec- disease exhaust the current conventional treat-
ondary to tumor growth, (2) fractures (both ment options and have persistent poorly con-
micro- and macro-), (3) cytokine-mediated trolled pain. Because of the shortcomings of the
osteoclastic bony destruction that results in currently available therapies, additional treat-
stimulation of nerve endings in the endosteum ment options for palliation of focal pain due to
(5-11), and (4) tumor growth into surrounding metastatic disease would be helpful. Fortu-
nerves and tissues. nately, many investigators currently are explor-
External-beam radiation therapy (RT) is the ing alternative therapies. Described below are
standard of care for patients who present with several reports that offer new image-guided
localized bone pain. This treatment results in percutaneous treatments for the palliation of
a reduction in pain for the majority of these pain from metastatic skeletal disease.

377
378 M.R. Callstrom et al

Percutaneous Therapies for procedure in 60 to 90 minutes. Three months


posttreatment, the patients had 30% to 45%
Palliation of Painful Metastases pain reduction.
Several new strategies recently have been
reported for the treatment of painful meta- Percutaneous Radiofrequency
static disease. All of these new methods are Ablation
based on using percutaneous image-guided
methods to deliver tissue-ablative devices into Recently, several case reports have appeared
focal metastatic lesions. These methods include that describe the treatment of painful meta-
the use of ethanol, laser-induced interstitial static lesions with percutaneous RFA. Included
thermotherapy (LITT), and percutaneous below are descriptions of several of these case
radiofrequency ablation (RFA). reports. Following this, we include results of an
ongoing prospective clinical trial designed to
determine the clinical magnitude and durabil-
Percutaneous Ethanol Administration ity of the use of RFA for the treatment of
painful metastatic bone disease.
Gangi and coworkers (18) described the use of
computed tomography (CT)-guided percuta-
neous administration of 95% ethanol for the Case Reports
palliation of pain from 27 metastatic bone As a prelude to treatment of spinal metastatic
lesions in 25 patients previously treated with disease, Dupuy and coworkers (20) used a pig
radiotherapy and/or chemotherapy. Sixteen model to examine the temperature distribution
lesions received a single dose of ethanol, while within a vertebral body and the adjacent spinal
10 lesions received two doses and one lesion canal with the RFA electrode placed within the
received three doses. The response of these vertebral body. They found decreased heat
patients to this treatment was assessed by the transmission in cancellous bone and an insula-
reduction in use of analgesic medicines 48 tive effect of cortical bone. Importantly, tem-
hours and 2 weeks following therapy. Complete perature elevations in the epidural space were
relief of pain was achieved in four patients, and not high enough to cause injuries to the adja-
very good but incomplete relief (75% analgesic cent spinal cord. Subsequently, these workers
medicine reduction) in 11 patients. Seven reported the treatment of a woman with
patients received little or no relief with the a painful osteolytic focal metastatic heman-
treatment. giopericytoma lesion in an anterior lumbar ver-
tebral body. Using local anesthesia and
conscious sedation, the lesion was accessed
Percutaneous Laser-Induced
from a lateral approach that passed through
Interstitial Thermotherapy
intact cortex with a 14-gauge Ackermann bone
Groenemeyer and coworkers (19) reported the biopsy needle (Cook Inc., Bloomington, IN).
treatment of three patients with spinal metas- Subsequently a 3-cm exposed tip Radionics
tases using a neodymium: yttrium-aluminum- radiofrequency electrode (Tyco Healthcare
garnet (Nd:YAG) laser with a wavelength of Group LP, Burlington, MA) was used to treat
1064 and a 400-lim fiber. With local anesthesia the lesion. The patient had improved pain
and CT guidance, a coaxial system was used via control at 13-month follow-up.
a transpedicular approach. Laser energy was Groenemeyer and coworkers (21) reported
applied at a power of 4 to 10 W with a pulse the RFA treatment of 10 patients with 21 unre-
length of 0.1 to 1.0 seconds at I-second inter- sectable painful spinal metastases using an
vals. To achieve coverage greater than 7 mm, the expandable-type electrode (RITA Medical
fiber was repositioned and the thermal treat- System, Inc., Mountain View, CA) and a 50-W
ment was repeated, allowing completion of the generator. The patients were treated under
30. Image-Guided Palliation of Painful Skeletal Metastases 379

local anesthesia only. Target temperature for dure was safe and resulted in significant relief
the ablations was based both on the distance of pain; therefore, the study was expanded to
from the spinal cord and patient tolerance for enroll patients from other centers in the United
the procedure. Four patients also were treated States and Europe. Here is a brief description
with vertebroplasty and received 3 to 5.5mL of of this ongoing prospective trial:
polymethylmethacrylate 3 to 7 days following
RFA. At last follow-up, nine of the 10 patients
Method
reported reduced pain, with an average pain
reduction of 74%. In an ongoing prospective trial, we have treated
Wood's group (22) reported the treatment of 43 patients at five centers in the United States
a patient with metastatic fallopian tube car- and Europe over the past two years. Patients
cinoma with multiple painful subcutaneous who were included had ;;::4/10 worst pain over a
masses. Using conscious sedation and local 24-hour period from :S;2 painful sites of metas-
anesthesia, the lesions were treated for 10 tases based on the Brief Pain Inventory (BPI)
minutes at a target temperature of 110°C with (28, 29). In the BPI, patients are asked to rate
a model 70 electrode and 50-W generator. their worst, least, and average pain in the past
Immediately following treatment, the patient 24 hours, with allowed responses ranging from
had no pain at the treated sites. She reported oto 10 (0 = no pain, 10 =pain as bad as you can
1-3/10 pain 1 month later. imagine). Relief of pain secondary to the RFA
Ohhigashi and coworkers (23) reported the procedure or to pain medications is scored on
RFA treatment of painful recurrent rectal a scale of 0% (no relief) to 100% (complete
cancer in the pelvis. Notably, recurrent rectal relief). Pain interference with daily living is
cancer may produce local pain, rectal bleeding, evaluated with questions concerning general
or bowel obstruction with resultant poor activity, mood, walking ability, normal work,
quality of life. Although a curative approach relations with other people, sleep, and enjoy-
such as total pelvic exenteration is possible, few ment of life, also on a 0 to 10 scale (0 = no inter-
patients are eligible (24). Unfortunately, radia- ference, 10 = completely interference).
tion therapy is palliative only and of limited Patients were treated under conscious seda-
clinical benefit (25). These workers treated two tion or general anesthesia at the discretion of
patients with 4-cm and 6-cm diameter recurrent the individual investigator. We have found that
rectal carcinomas located anterior to the an epidural catheter, placed prior to treatment
sacrum that had not responded to chemo- of lesions, greatly improves patient pain control
therapy or radiation therapy. Using epidural following the procedure when the lesion is
catheter-mediated analgesia, a LeVeen RF located in the pelvis or lower extremities. Two
electrode (Boston Scientific Corp., Natick dispersive electrodes (grounding pads) are
MA.), was placed into the lesions using CT placed on the patient at equidistant sites from
guidance. Both patients reported a decrease in the RF source, usually on the patient's thighs.
pain following the treatment and a reduction in To avoid skin burns from the ground pads, we
oral analgesic requirements. place two monitoring temperature electrodes
(Mallinckrodt Mon-a-therm Model 4070 with
700 series thermistor) on the corners of the
Clinical Trial
leading edges (nearest the ablation location) of
Because of several reports that suggest the both grounding pads. If the skin temperature
potential benefits of RFA for palliation of pain reaches 38°C, dry cold packs are applied over
due to metastatic disease, we conducted a fea- the grounding pads. All patients included in the
sibility clinical trial to determine the safety and study were treated with a Starburst XL model
benefits of RFA in patients with painful needle (RITA Medical Systems, Mountain
metastatic lesions that involve bone (26, 27). View, CA), a 14-gauge/6.4-Fr device, with an
Our preliminary data showed that this proce- active electrode trocar tip, and nine electrodes
380 M.R. Callstrom et al

A-C
FIGURE 30.1. (A) Radiofrequency ablation (RFA) computed tomography (CT) guidance. (C) CT image
electrode with full deployment (Starburst XL elec- with RFA electrode deployed in an osteolytic lesion
trode, RITA Medical Systems). (B) Percutaneous in the acetabular region.
introduction of RFA electrode using intermittent

spread in a ball-like fashion that generate up to involved in the destructive metastatic lesion.
a 5-cm-diameter zone of necrosis. Electrode deployment diameters are chosen to
A single ablation typically is performed for adequately cover the metastatic lesion without
lesions of ~3 cm in diameter with a target tem- damaging nearby normal, uninvolved tissues.
perature of lOO°C maintained for a minimum In other centers, some lesions have been
of 5 minutes. For larger lesions, multiple sys- treated with placement of the electrode into the
tematic deployments are used with 3- to 5-cm center of the lesion with resultant debulking of
deployments of the electrode with the lOO°C the lesion, but with incomplete ablation of the
target temperature maintained for 5 to 10 soft tissue/bone interface. Figure 30.3 shows a
minutes. For lesions >5 cm in diameter, the large osteolytic metastatic lesion involving the
entire lesion was often not completely treated; sacrum 6 weeks following RFA. This image
rather, the ablation treatments are focused on shows central low attenuation within the
the margin of the bony lesion, with the goal of
treating the soft tissue/bone interface. For full
deployment of the electrode, a target tempera-
ture of lOO°C was maintained for 5 to 15
minutes.
Figure 30.1 shows the needle fully deployed,
typical CT-guided placement of the needle, and
deployment of the tines into an osteolytic
lesion in the periacetabular region. This figure
illustrates that the electrode is advanced into
the soft tissue portion of the lesion to be
treated, and the tips of the tines of the electrode
are placed against the soft tissue/bone inter-
face. Figure 30.2 shows another example of an
osteolytic lesion that involves the caudal aspect
FIGURE 30.2. RFA treatment of the bone-tumor
of the sacrum with associated partial destruc- interface. Prone CT demonstrates an osteolytic mass
tion. The RF electrode is placed with the tines involving the left caudal aspect of the sacrum. This
of the deployed electrode driven against the mass causes destruction of the adjacent bone. The
soft tissue/bone interface. Subsequent elec- bone-tumor interface is a key site for electrode
trode deployments (not shown) are performed deployment to destroy nerve endings that were the
to completely treat the portion of bone likely cause of pain.
30. Image-Guided Palliation of Painful Skeletal Metastases 381

pubic bones with osteolytic destruction, most


marked on the left. On examination, the patient
described 8/10 pain from the left pubic bone
lesion with minimal pain to the right of midline.
The left pubic bone lesion was treated with
several overlapping deployments. Four weeks
following treatment, the patient's pain dropped
from 8/10 to 0110. The patient died 12 weeks
posttreatment with 3/10 pain at the treated site.
We have treated several patients with recur-
rent or residual metastatic rectal carcinoma
centered anterior to the sacrum in the presacral
FIGURE 30.3. Failed palliation of pain due to RFA space, often with associated osteolytic destruc-
treatment of the central portion of a metastatic tion of the underlying sacrum. Figure 30.6
lesion without treatment of the tumor-bone inter- shows a typical lesion in a 38-year-old man that
face. CT performed 6 weeks post-RFA demonstrates measured approximately 3 em in diameter and
necrosis in the treated central portion of the tumor was located at the level of the coccyx with 8/10
with moderate residual tumor that involves the pain. The patient's pain was reduced to 2/10 at
sacrum and iliac bone. To be effective, the RFA treat- week 4, and the patient reports 1/10 pain at the
ment should be at the tumor-bone interface. treated site at the latest follow-up interview,
Debulking of the central portion of metastatic
11 months following treatment.
lesions is not effective to reduce pain.

metastatic lesion consistent with necrosis from


the RFA procedure. Residual tumor is clearly
present along the medial aspect of the sacrum
and against the adjacent iliac bone. Following
this treatment, the patient failed to derive
reduction in pain and had no clear benefit from
the procedure.
We have found that it is critically important
to examine each patient prior to the RFA treat-
ment to determine if the patient's pain corre-
sponds to an identifiable lesion on CT, magnetic
resonance imaging (MRI), or ultrasound (US)
imaging. Figure 30.4 shows a prone CT image
of a metastatic osteolytic lesion that involves
the mid sacrum. We examined the patient and
placed a metallic marker on the skin overlying
the site of greatest pain. As shown in Figure
30.4 the site of pain corresponded to a sacral FIGURE 30.4. Pain due to sacral fracture rather than
osteolytic tumor. Prior to prone CT examination, the
fracture on the side opposite of the destructive
patient was examined and a metallic marker was
sacral lesion. Because the patient'S pain likely
placed on the skin overlying the site of focal pain.
resulted from the underlying sacral fracture, we CT imaging at this level shows an osteolytic destruc-
did not treat the adjacent destructive sacral tive lesion involving the right side of the sacrum. The
lesion. metallic marker (arrowhead) overlies a left sacral
Figure 30.5 shows CT images of the pelvis of fracture (arrow). This lesion was not treated because
a woman with metastatic endometrial carci- the patient's pain was due to the sacral fracture
noma. A large destructive lesion involves both rather than the adjacent osteolytic lesion.
382 M.R. Callstrom et al

ment included resection of the rectum with a


diverting colostomy. The patient had reduced
bladder function with difficulty initiating void-
ing and incomplete emptying of his bladder.
Because of the desire to retain the patient's
bladder function, we systematically treated the
lesion in two stages, 6 weeks apart, with a total
of 14 electrode deployments; the initial treat-
ment focused on the caudal aspect of the
sacrum. Figure 30.7B shows one of the seven
RFA electrode deployments in the first stage of
the treatment. Following the initial treatment,
the patient was able to sit in bed the same day,
A

B
A
FIGURE 30.5. RFA of a pubic symphysis metastasis.
(A) CT scan demonstrates osteolytic destructive
lesions involving both pubic bones due to metastatic
endometrial carcinoma. The lesion on the patient's
left caused severe pain in spite of prior treatment
with chemotherapy and radiation therapy. (B) RF
electrode deployed in the osteolytic lesion. Patient's
pain decreased from 8110 before RF ablation to 3/10
4 weeks posttreatment.

Large painful lesions may be treated effec-


tively with RFA. Figure 30.7 shows a portion of B
a destructive infiltrative metastatic rectal carci-
FIGURE 30.6. Metastatic presacral rectal carcinoma.
noma lesion that involves the sacrum. This
(A) Prone contrast-enhanced CT demonstrates an
lesion measured approximately 11 cm in diam-
oval peripherally enhancing soft tissue mass anterior
eter and involved a majority of the sacrum. to the coccyx (arrows). (B) Prone CT demonstrates
Figure 30.7A is a CT image at the level of the the RFA electrode deployed within the mass.
lower sacrum that shows the large destructive Patient's pain decreased from 8/10 to 1/10 4 weeks
lesion. This patient was unable to sit or lie on posttreatment. The patient continues to report 1/10
his back due to severe pain (8/10). Prior treat- pain at this site 11 months posttreatment.
30. Image-Guided Palliation of Painful Skeletal Metastases 383

A B
FIGURE 30.7. RFA of a large sacral metastasis. (A) aspect of the osteolytic lesion. The second stage was
Prone CT demonstrates near-complete replacement performed 6 weeks later. Patient was unable to sit
of the sacrum by metastatic rectal carcinoma. (B) prior to treatment and had resting pain of 8/10. One
RFA electrode deployed to Scm. This was one of day following the ablation, the patient was able to sit.
seven electrode placements during the first stage of His pain decreased to 3/10.
treatment for the ablation of much of the caudal

and described 3/10 pain in the treated region 4 the treatment to 3110 4 weeks after the RFA
weeks following the procedure. Because of the procedure.
patient's desire for greater pain relief, and with Immediate postprocedural pain has been
the understanding that further RFA could lead treated with epidural or intravenous (IV)
to loss of bladder function, we treated the supe- opioid analgesics. Depending on the size of
rior portion of the lesion up to the level of the the treated lesion and the degree of post-
52 neural foramina. Within 4 weeks, his pain operative pain, patients typically were observed
was reduced to 0110 at the treated region. For- overnight in the hospital. Patients with per-
tunately, his bladder function was not disturbed sistent postprocedural pain were administered
by the treatment. oral opioid analgesics at the time of discharge.
Radiofrequency ablation of painful para-
spinal metastatic lesions also is possible.
Extreme caution must be used when metastatic
Pain Assessment and Follow-Up
lesions have destroyed the vertebral body with
resultant loss of the insulative effect of the adja- The patient's pain was the primary end point
cent bone (20). Figure 30.8A is a CT image of and was measured using the BPI. We inter-
a large metastatic colorectal carcinoma lesion viewed all patients with the BPI just prior to the
that involves a rib, vertebral body, and pleural procedure, the day following the treatment,
surface. To avoid thermal damage to the spinal weekly for 1 month, and then every 2 weeks for
cord, we placed a passive thermocouple along the second through the sixth month. Each
the lateral aspect of the destroyed pedicle (Fig. patient was asked to answer questions with
30.8B,C). We subsequently treated the lesion respect to the lesion that was treated.
with three electrode deployments along the The type of malignancy, size, and location of
involved rib. With the electrode deployed in the the lesions that were treated are summarized in
nearest paraspinal location (Fig. 30.8D), we Table 30.1 Colorectal (n = 10), renal (n =9), and
discontinued further treatment when the pas- lung (n = 4) were the most common tumor
sive thermocouple reached 40°C. This patient types treated. The most common sites of tumor
reported a reduction in pain from 10/10 prior to involvement were the pelvis (n = 12), sacrum
384 M.R. Callstrom et al

A
c

B D
FIGURE 30.8. Metastatic colorectal carcinoma to rib, in the metastatic lesion. (C) Prone CT demonstrates
vertebral body, and pleural surface. (A) Volumetric the most medial deployment of the RFA electrode
prone CT demonstrates osteolytic destruction of the (arrow). Ablation was discontinued when the tem-
lateral portion of the vertebral body (arrow) with perature at the passive thermocouple reached 40°C.
associated soft tissue mass anterior to the rib. (B) (D) Photograph of the RFA electrode in place with
Prone CT image shows the position of the passive adjacent thermocouple located medially. The
thermocouple probe near the vertebral body pedicle patient's pain decreased from 10/10 to 3/10 4 weeks
with the RFA electrode deployed laterally (arrow) following the treatment.

(n = 12), rib (n = 6), and vertebrae (n = 4). Pain Response to Radiofrequency Ablation
Treated lesions were osteolytic, with the excep-
tion of two patients who had mixed osteolytic! A total of 41/43 patients (95%) experienced a
osteoblastic lesions. The size of the treated drop in pain that met our a priori definition of
lesions ranged from 1 cm (rib) to approximately a clinically significant benefit (two-point drop).
18cm (paraspinal). The median number of abla- Patients experienced highly significant reduc-
tions per lesion was 3.0 (range 1-14), with an tions in worst pain, average pain, pain inter-
average time per ablation of 11.7 minutes ference, and improvement in pain relief after
(range 1.1-52.5 minutes). The mean total RFA of painful metastases involving bone (Fig.
ablation time was 49.5 minutes (range 8.0- 30.9 and Table 30.2). Significant decreases were
218.9 minutes). seen beginning at week 1 and extending to week
30. Image-Guided Palliation of Painful Skeletal Metastases 385

TABLE 30.1. Characteristics of patients treated with 24 for all pain parameters. Two patients, whose
radiofrequency ablation (RFA). pain responded to initial RFA, required retreat-
Number of patients 43 ment after a recurrence of pain (:?:4/10 worst
Women 15 (35%) pain) at 8 and 16 weeks, respectively. Both
Men 28 (65%) patients had significant reduction in worst pain
Age (median) 64 (range 28-88) (:?:4-point drop) following retreatment.
Thmor type (number)
Colorectal carcinoma 10
Opioid use was recorded for each patient and
Renal carcinoma 9 converted into morphine equivalents using
Lung carcinoma 4 standardized conversions (30). Following RFA,
Sarcoma 3 opioid requirements peaked at week 1 (Table
Other 17 30.2). At week 4, although opioid requirements
Tumor size (longest 6.3 em (range 1.4-18.0)
diameter; cm)*
were not statistically different from baseline, a
Tumor location trend toward decreasing requirements was'
Pelvis 12 seen. By weeks 8 and 12, there were significant
Sacrum 12 reductions in opioid usage. Increases in opioid
Rib 6 usage occurred at week 24, although the corre-
Vertebrae 4
Other 9
sponding pain scores did not increase.
Prior radiation to treated site 32 (74%) Although the RFA procedure is safe, pre-
Concurrent opioid analgesics 30 (70%) cautions are necessary to avoid thermal injury
to large nerves, vessels, bowel, and bladder.
* Tumor size at presentation. For lesions >5 em, RFA treat-
ment targeted ablation of the tumor-bone interface, not
Adverse events following the procedures were
complete ablation of the tumor. noted in three patients. One patient developed

A B
10 10
c
c '[ 8
'[ 8 Gl

ii 8 ~ 8
~ 4
c ~ 4
i
c
2 :ll 2
::IE
Oir-r........-....._.__.....,...-_........._____ O.....................................---.--__~
Week 0 1 2 348 8 12 18 20 24 Week0123488 12 18 20 24
n- 43424142413633 26 19 14 12 n- 43424142413633 26 19 14 12
c D
100 ~ 10
c
:! 80 l!! 8
f ~
c80 S 8
·fti .5
~40 .[ 4
l'lI
~ 20 c 2
:ll
0 .......,...,...................--.-_....-......._ ... 0 ..............................- _.......-
::IE ...........0\
Week 0 1 2 348 8 12 18 20 24 Week 0123488 12 18 20 24
n= 43414040403328 21 17 12 12 n- 43424041413633 26 19 14 12

FIGURE 30.9. Mean Brief Pain Inventory (BPI) pain daily activities. Error bars represent the 95% confi-
scores over time for patients treated with RFA. (A) dence intervals. n, the number of patients complet-
Worst pain. (B) Average pain. (C) Pain relief from ing BPI at each time point.
RFA or medications. (D) Interference of pain in
386 M.R. Callstrom et al

TABLE 30.2. Number of patients, Brief Pain Inventory (BPI) mean pain scores, and opioid requirements at
baseline and following RFA.
Baseline Day 1 Week 1 Week 4 Week 8 Week 12 Week 24

No. of patients 43 31* 42 41 33 26 12


Worst pain 7.9 7.3 5.8 4.5 3.5 3.0 1.4
(0-10) P = .2456 P < .0001 P < .0001 p < .0001 P < .0001 p = .0005
Average pain 5.8 4.6 3.9 2.8 2.4 1.9 1.2
(0-10) P < .0001 p < .0001 P < .0001 p < .0001 p < .0001 P = .0005
Pain interference 6.6 5.6 5.0 3.7 3.5 2.9 1.3
(0-10) P = .0825 p = .0001 p < .0001 p < .0001 p = .0008 p = .0015
Pain relief 43 64 69 73 74 79 84
(0-100) P = .0007 p < .0001 P < .0001 p = .0002 P < .0001 p = .0029
Morphine 99.0 97.5 105.7 95.5 40.4 45.4 93.0
equivalent dose p = .01 p=.01 P =.47

Note: p values are signed rank tests of the null hypothesis that the difference in the current time period minus the
baseline value is equal to zero.
* Ultrasound cohort only.

a second-degree skin burn at the grounding pad Another patient sustained an acetabular frac-
site. Another patient developed transient bowel ture 6 weeks following RFA of a metastatic
and bladder incontinence following RFA of a lesion involving the ileum, ischium, and acetab-
previously irradiated leiomyosarcoma metasta- ulum. It was unclear whether RFA contributed
sis that involved the upper sacrum. A third to the development of the acetabular fracture.
patient developed an acetabular fracture 6 It is possible that injection of polymethyl-
weeks following RFA of a breast cancer metas- methacrylate into the ablated tissue, as per-
tasis with significant involvement of the ileum, formed by Groenemeyer and coworkers (21)
ischium, and acetabulum. This patient required following ablation of spinal metastases, could
open reduction and fixation of the acetabulum. result in stabilization of the partially destroyed
bone and prevent fracture. One patient suf-
fered a second-degree skin burn at the ground-
Summary ing pad site. Further burns may be prevented
through careful monitoring of skin temperature
Severaf case reports and a clinical trial have deep to the grounding pads.
found a highly significant reduction in pain Radiofrequency ablation provides an effec-
from metastatic disease with percutaneous tive, rapid, and durable method for palliation of
ablation. These findings are important, not only localized painful metastases that involve bone.
because of the magnitude of the benefit, but This procedure is safe and the relief of pain is
because it occurred in patients traditionally significant; the procedure should provide an
refractory to most conventional treatments. alternative for the palliation of painful bone
These reports and our clinical trial show that metastases when standard treatments fail.
RFA of metastatic lesions that involve bone is
a safe procedure. Three complications were
noted in our trial following treatment with References
RFA. One patient suffered transient bowell 1. Nielsen OS, Munro AJ, Tannock IF. Bone metas-
bladder incontinence following treatment of a tases: pathophysiology and management policy.
sacral metastasis that was previously irradiated. J Clin OncoI1991;9:509-524.
30. Image-Guided Palliation of Painful Skeletal Metastases 387

2. Mercadante S. Malignant bone pain: patho- 15. Price P, Hoskin PJ, Easton D. Prospective ran-
physiology and treatment. Pain 1997;69:1-18. domised trial of single and multifraction radio-
3. Mantyh PW, Clohisy DR, Koltzenburg M, Hunt therapy schedules in the treatment of painful
SP. Molecular mechanisms of cancer pain. Nat bony metastases. Radiother Oncol 1986;6:247-
Rev Cancer 2002;2:201-209. 255.
4. Mannion RJ, Woolf CJ. Pain mechanisms and 16. Cole DJ. A randomized trial of a single treat-
management: a central perspective. Clin J Pain ment versus conventional fractionation in the
2000;16:S144-S156. palliative radiotherapy of painful bone metas-
5. Watkins LR, Goehler LE, ReIton J, Brewer MT, tases. Clin Oncol (R ColI Radiol) 1989;1:59--62.
Maier SF. Mechanisms of tumor necrosis factor- 17. Gaze MN, Kelly CG, Kerr GR, et al. Pain relief
alpha (TNF-alpha) hyperalgesia. Brain Res 1995; and quality of life following radiotherapy for
692:244-250. bone metastases: a randomised trial of two
6. Sorkin LS, Xiao WH, Wagner R, Myers RR. fractionation schedules. Radiother Oncol 1997:
Tumour necrosis factor-alpha induces ectopic 109-116.
activity in nociceptive primary afferent fibres. 18. Gangi A, Kastler B, Klinkert A, Dietemann JL.
Neuroscience 1997;81:255-262. Injection of alcohol into bone metastases under
7. Woolf CJ, Allchorne A, Safieh-Garabedian B, CT guidance. J Comput Assist Tomogr 1999;18:
Poole S. Cytokines, nerve growth factor and 932-935.
inflammatory hyperalgesia: the contribution of 19. Groenemeyer DH, Schirp S, Gevargez A. Image-
tumour necrosis factor alpha. Br J Pharmacol guided percutaneous thermal ablation of bone
1997;121:417-424. tumors. Acad RadioI2002:467-477.
8. Opree A, Kress M. Involvement of the proin- 20. Dupuy DE, Hong R, Oliver B, Goldberg SN.
flammatory cytokines tumor necrosis factor- Radiofrequency ablation of spinal tumors: tem-
alpha, IL-1 beta, and IL-6 but not IL-8 in the perature distribution in the spinal canal. AJR
development of heat hyperalgesia: effects on 2000;175:1263-1266.
heat-evoked calcitonin gene-related peptide 21. Groenemeyer DHW, Schirp S, Gevargez A.
release from rat skin. J Neurosci 2000;20: Image-guided radiofrequency ablation of spinal
6289-6293. tumors: preliminary experience with an expand-
9. Davar G, Hans G, Fareed MU, Sinnott C, able array electrode. Cancer J 2002;8:33-39.
Strichartz G. Behavioral signs of acute pain pro- 22. Patti JW, Neeman Z, Wood BJ. Radiofrequency
duced by application of endothelin-1 to rat ablation for cancer-associated pain. J Pain 2002;
sciatic nerve. Neuroreport 1998;9:2279-2283. 3:471-473.
10. Gokin AP, Fareed MU, Pan HL, Hans G, 23. Ohhigashi S, Nishio T, Watanabe F, Matsusako
Strichartz GR, Davar G. Local injection of M. Experience with radiofrequency ablation in
endothelin-l produces pain-like behavior and the treatment of pelvic recurrence in rectal
excitation of nociceptors in rats. J Neurosci cancer; report of two cases. Dis Colon Rectum
2001;21:5358-5366. 2001;44:741-745.
11. Zhou Z, Davar G, Strichartz G. Endothelin-1 24. Ogunbiyi OA, McKenna K, Birnbaum EH,
(ET-l) selectively enhances the activation gating Fleshman JW, Kodner IJ. Aggressive surgical
of slowly inactivating tetrodotoxin-resistant management of recurrent rectal cancer-is it
sodium currents in rat sensory neurons: a mech- worthwhile? Dis Colon Rectum 1997;40:150-
anism for the pain-inducing actions of ET-1. 155.
J Neurosci 2002;22:6325-6330. 25. Wong CS, Cummings BJ, Brierley JD, et
12. Massie MJ, Holland Je. The cancer patient with al. Treatment of locally recurrent rectal
pain: psychiatric complications and their man- carcinoma-results and prognostic factors. Int J
agement. J Pain Symptom Manag 1992;7:99-109. Radiat Oncol BioI Phys 1998;40:427-435.
13. Spiegel D, Sands S, Koopman e. Pain and 26. Callstrom MR, Charboneau JW, Goetz MP, et al.
depression in patients with cancer. Cancer 1994; Painful metastases involving bone: feasibility of
74:2570-2578. percutaneous CT- and US-guided radio-
14. Jeremic B, Shibamoto Y, Acimovic L, et al. A frequency ablation. Radiology 2002;224:87-97.
randomized trial of three single-dose radiation 27. Goetz M, Rubin J, Callstrom M, et al. Per-
therapy regimens in the treatment of metastatic cutaneous US and CT-guided radiofrequency
bone pain. Int J Radiat Oncol BioI Phys 1998; ablation of painful metastases involving bone.
42:161-167. ASCO 2002; May 24-30:1544.
388 M.R. Callstrom et al

28. Daut RL, Cleeland CS, Flanery RC. Develop- 30. Farrar JT, Young JP Jr, LaMoreaux L, Werth JL,
ment of the Wisconsin Brief Pain Questionnaire Poole RM. Clinical importance of changes in
to assess pain in cancer and other diseases. Pain chronic pain intensity measured on an ll-point
1983;17:197-210. numerical pain rating scale. Pain 2001;94:149-
29. Cleeland CS, Gonin R, Hatfield AK, et al. Pain 158.
and its treatment in outpatients with metastatic
cancer. N Engl J Med 1994;330:592-596.
31
Radiofrequency Ablation of
Osteoid Osteoma
Daniel I. Rosenthal and Hugue Ouellette

Percutaneous radiofrequency treatment of Malignant transformation of an osteoid osteoma


osteoid osteoma has been performed for more has not been described.
than 10 years. It is the preferred method of Osteoid osteomas comprise approximately
treatment for most patients in those centers 10% of benign bone tumors (8). They are thus
that offer it, and has been shown to be a safe, rare enough so that the physician who initially
effective, and cost-effective alternative to oper- sees the patient may fail to consider the diag-
ative treatment (1-5). However, many patients nosis, but common enough to be encountered
continue to be subjected to the increased risks in many practices.
and lengthy recovery of open surgery or to the
prolonged discomfort and risks associated with
medical therapy. This is presumably because of
Patient Identification
lack of awareness of the technique or benefits
of percutaneous treatment. This chapter pro-
Clinical
vides a detailed description of proper patient The tumors are usually found in the extremities
selection, technique, and follow-up for those of children and young adults. The typical patient
wishing to perform this therapy. is a late teenage boy. The average age in our
series is 17, with a male predominance of 3: 1.
Patients less than 5 years of age or over 30 are
Definition uncommon, although a scattering of very young
and very old patients have been reported (9,10).
Osteoid osteoma is a small, benign, but painful Osteoid osteomas are painful. They appear to
bone-forming tumor, which may cause con- cause pain by two mechanisms. The lesions
siderable peritumoral inflammation. It is produce a large quantity of prostaglandin,
composed of osteoblasts that give rise to leading to a variable but often marked inflam-
variable amounts of woven bone and a loose mation of surrounding tissues (11,12). In this
fibrovascular stroma (6). Unlike osteoblas- respect, they are similar to chondroblastomas,
toma, which may have identical histologic fea- which also produce prostaglandin (12). In addi-
tures, osteoid osteoma has little or no growth tion, however, osteoid osteomas contain
potential. The nonprogressive nature of the sensory nerves (11,13). The combination of
disease is a key aspect of the definition (7). these two features is, we believe, unique among
However, since many, or even most, lesions are bone tumors (14).
not observed over time to determine growth, in The pain often begins insidiously as a dull
practice, osteoid osteoma is often differentiated ache in the affected part. Because of its char-
from osteoblastoma on the basis of size acteristic occurrence in young males, it is often
(osteoid osteoma being smaller than 1.5 cm). ascribed to an athletic injury or to "growing

389
390 D.I. Rosenthal and H. Ouellette

pains". With time, the pain may become sharper smaller than the reactive zone that surrounds it.
and more severe. Although pain may be exac- Typically, osteoid osteoma is an oval or spheri-
erbated by activity, the tumors are usually cal lucency, measuring between 1 and 15 mm in
painful at rest, and are often most painful at diameter (average 7-8mm) (Figs. 31.1 to 31.3).
night. This is a useful distinguishing feature Occasionally, tumors may be markedly elon-
from stress fracture, which may have similar gated, measuring several centimeters in longi-
imaging features, but does not hurt when the tudinal dimension, but less than 1 cm in
extremity is rested. Superficial lesions are quite transverse diameter (16) (Fig. 31.4). This varia-
sensitive to palpation. tion is most often seen in young children.
Pain usually responds to nonsteroidal anti- Rarely, a small cluster of tumors may be seen
inflammatory medications. Response to med- (Fig. 31.5). Such "multicentric" tumors are
ication is often (but not always) rapid and almost always confined to a small anatomic
complete. Aspirin and ibuprofen appear to area within a single bone (17). Failure to rec-
provide symptomatic relief in the greatest ognize that multiple lesions are present may
number of individuals. Naproxen, if effective, be an uncommon cause of recurrence (18).
offers the greatest duration of relief. Acet- Involvement of two anatomically remote sites
aminophen is less effective. Severe pain may has been reported (19-21), but is exceedingly
be accompanied by marked muscle wasting, rare.
sometimes simulating the presentation of a The tumor may contain a variable amount of
neuropathy (15). internal ossification, but is rarely completely
ossified (Figs. 31.1 and 31.2). The term nidus
often is used to describe the appearance of
Imaging osteoid osteoma; however, it is sometimes used
For therapy to be appropriately directed, the to mean the lucent tumor within the zone of
tumor must be definitively identified by pre- reactive sclerosis, and at other times is used to
procedure imaging studies. This can be chal- refer to the focal ossification within the tumor.
lenging since the tumor frequently is much The word nidus comes from the Latin term for

A B
FIGURE 31.1. (A) Plain film of the femur demon- form. (B) Computed tomography (CT) scan through
strates a lucency within the posterior cortex of the the lesion demonstrates that the tumor is 8mm in
femur,surrounded by dense periosteal new bone for- diameter and contains faint internal ossification
mation (arrow). This is the typical appearance of an (arrow).
intracortical osteoid osteoma, the most common
A B

FIGURE 31.2. (A) Plain film demonstrates a lesion with a very similar appearance to that in Fig. 31.1A. (B)
CT scan shows that the lesion originated on the periosteal surface of the bone, another common site of origin.

A B
FIGURE 31.3. (A) The lesion (arrow) is difficult to see on this plain radiograph. (B) CT scan shows that the
tumor has an intramedullary location, the least common presentation.

FIGURE 31.4. Reformatted CT scans demonstrate an


elongated osteoid osteoma in the cortex of the
FIGURE 31.5. Reformatted CT scans demonstrate a
femur. The tumor measured almost 3cm in longitu-
multicentric osteoid osteoma of the femur, showing
dinal dimension, but only 8mm in axial diameter.
two distinct tumors separated by intervening bone
(arrow).
392 D.I. Rosenthal and H. Ouellette

A B
FIGURE 31.6. Sagittal (A) and axial (B) magnetic osteoma. The tumor itself is difficult to see, but
resonance images of the elbow demonstrate a large appears as a rounded low-signal area in the humeral
joint effusion in response to an intraarticular osteoid condyle (B, arrow).

"nest," and perhaps would be most suitable to may produce a massive joint effusion, simulat-
describe the lucent tumor, with the tumoral ing an arthropathy (Fig. 31.6) (22). Solid or
ossification corresponding to the "egg" in the sometimes layered periosteal ossification and
nest. However, in our opinion, the term is medullary sclerosis are common. The sclerotic
ambiguous. Throughout this chapter we refer to bone that surrounds the tumor can be exten-
"tumor" and "tumor ossification," avoiding the sive, and can obscure the tumor on plain films.
word nidus. Computed tomography (CT) scan reveals the
Osteoid osteomas arise in cortex and perios- diagnostic characteristics of osteoid osteoma
teum with equal frequency. Medullary tumors most reliably. Magnetic resonance imaging
are much less common (Fig. 31.3). (MRI) is highly sensitive to the reactive tissues,
Although any bone may be affected, tumors including the bone marrow and soft tissue
are most common in the lower extremities. edema that often surround the tumor. How-
Local pain usually directs imaging studies to ever, the tumor itself may be more difficult to
the appropriate location. However, referred identify on MRI (Fig. 31.6) (23). If the charac-
pain is not rare, especially in younger patients. teristic round or oval tumor cannot be identi-
The most common pattern of referred pain is a fied with imaging studies, radiofrequency treat-
tumor of the hip resulting in knee symptoms. ment should not be attempted.
Radioisotope scans are reliably abnormal in
patients with osteoid osteoma. The tumor pro-
duces a small focus of intense uptake that is
apparent even in the presence of surrounding Other Lesions that May
tissue reaction. This feature can be useful to Appear Similar
identify the location of the abnormality and
sometimes may even be diagnostic. Several other lesions may cause confusion.
Osteoid osteomas are characteristically Other symptomatic benign bone tumors also
(although not universally) surrounded by may also result in small oval lucencies sur-
extensive peritumoral inflammatory changes. rounded by reactive bone. Painful chondromas
Bone marrow, periosteal, and soft tissue edema are usually periosteal, but rarely may be intra-
may be present. Lesions arising within joints cortical (24,25). Periosteal chondromas usually
31. Radiofrequency Ablation of Osteoid Osteoma 393

sit on the bone surface like an egg in a cup, sur- not be adequate therapy. If the correct diagno-
rounded by a base of periosteal bone, but not sis is revealed by the biopsy performed at the
by the extensive sclerosis seen with osteoid time of treatment, adjuvant therapy could be
osteoma (26). Chondroblastoma usually is performed with no adverse consequences for
found in the epiphyseal centers of growing the patient.
bones (as osteoid osteoma also may be). It is There are two entities for which radiofre-
usually larger than an osteoid osteoma, and quency treatment should be regarded as con-
may have a lobulated contour. The surrounding traindicated. A stress fracture would probably
tissue reaction may be very similar to osteoid be devitalized by radiofrequency treatment,
osteoma. Similarly, eosinophilic granuloma perhaps resulting in delayed healing. Stress
(EG) may cause marked marrow edema and fractures can be distinguished on imaging by
periosteal reaction. The lucent center of EG is the absence of the typical oval or spherical
often larger and more irregular in shape than lucency. A stress fracture may demonstrate a
osteoid osteoma (Fig. 31.7). central lytic area particularly in the tibia, but
In each of these cases, the typical lesion the lucency is usually less well defined, and
differs in appearance from the typical osteoid more linear. More importantly, the pain due to
osteoma, but atypical lesions may be indistin- a stress fracture improves with rest, and seldom
guishable. However, treatment of a small lesion disturbs the patient's sleep, while osteoid
of any of them by radiofrequency is appropri- osteoma generally causes pain that is not
ate and may be curative (27,28). activity-dependent.
The very rare intracortical variant of Treatment of an intracortical abscess by
osteosarcoma also may appear identical to an radiofrequency could result in formation of an
osteoid osteoma (29). Small metastases also area of devitalized bone. Such an iatrogenic
could have a similar appearance, but fortu- sequestrum could complicate efforts to treat
nately are extremely rare in the age group the infection. Most abscesses cause larger and
affected by osteoid osteoma. Radiofrequency less regular lytic areas, often resulting in
treatment of one of these malignant lesions has lobular, elongated lucencies through the scle-
not been reported, but would almost certainly rotic bone, resembling an animal's burrow.

A B
FIGURE 31.7. (A) Plain film showing a I-cm lytic scan of the lesion shows that the lesion has an irreg-
lesion of the femur, with marked periosteal reaction ular lobulated contour, which would be very unusual
on the lateral cortex. This lesion was originally for an osteoid osteoma. Biopsy confirmed the diag-
thought to represent an osteoid osteoma. (B) A CT nosis of eosiophilic granuloma.
394 D.l. Rosenthal and H. Ouellette

Although potentially an abscess may simulate If the untreated tumor is likely to result
an osteoid osteoma exactly (30), this must be in structural damage, intervention should be
quite uncommon, as we have not yet encoun- strongly considered. If the lesion is close to an
tered such a case. open growth plate, inflammatory hypervascu-
larity may cause overgrowth or premature
fusion leading to curvature of the limb or limb-
Rationale for Intervention in length inequality (38). Similarly, inflammatory
Osteoid Osteoma effects of intraarticular tumors may cause joint
damage that leads to late arthritis (39,40).
Since growth potential is limited, and since Spinal lesions often cause scoliosis due to
malignant degeneration has not been reported, muscle spasm. If left untreated for long periods
the primary goal of treatment is elimination of of time, this may become permanent (41).
symptoms. There is much anecdotal evidence,
and at least small series that suggests that these
lesions undergo spontaneous resolution, even if Choice of Intervention
untreated (31-33). The average time to resolu-
tion of symptoms is said to be from 3 (32) to 6 Although several effective percutaneous tech-
years (31). These observations have given rise niques exist, we favor radiofrequency treatment
to the belief that osteoid osteoma may be because of its effectiveness, low cost, and favor-
treated appropriately by nonsteroidal antiin- able complication rate. Laser treatment is also
flammatory drugs without surgical or percuta- a form of ablative therapy. It can be performed
neous intervention (32,34). with very small-caliber needles (42,43). How-
However, there is also anecdotal evidence of ever, in our experience this confers no advan-
symptoms persisting for much longer periods tage, as sturdy needles often are required to
without resolution (35-37). Since the majority gain access to the lesions and to obtain ade-
of patients appear to undergo intervention of quate biopsy material. Laser equipment is also
some sort, it is difficult to be certain of whether more costly. Percutaneous excision also can be
the majority of lesions would regress over effective (44-46), especially for smaller tumors.
time. When surgical treatment was the only Larger lesions are more difficult to remove,
alternative, chronic medical therapy was a and complications due to the extent of bone
reasonable alternative. However, percuta- removal may occur (47). Although success has
neous treatment has significantly lessened the been reported for alcohol injection (48,49),
morbidity of treatment, making it less clear why anatomic localization is more difficult to con-
one would elect to have long-term medical trol and ablative effects are less reliable.
therapy. Nonetheless, in considering the pros
and cons of percutaneous radiofrequency treat-
ment, all patients should be informed of this Contraindications to
option. Radiofrequency Ablation
There are several instances in which medical
treatment is not a good option. Some patients Complete diagnostic certainty is not required
do not tolerate nonsteroidal antiinflammatory prior to intervention, as other benign bone
medications. Gastrointestinal side effects are tumors may be treated without harm and with
relatively common; ulcer formation and bleed- possible benefit to the patient, if they are the
ing are uncommon. For a few patients, pain cause of pain. However, radiofrequency treat-
relief with nonsteroidal medications is inade- ment should not be performed if an infection
quate. If a patient is forced to alter his lifestyle, or a stress fracture is a strong consideration.
or is unable to sleep or requires narcotics for If the tumor cannot be identified within the
pain relief, intervention seems preferable to sclerotic bone and reactive edema, percuta-
conservative treatment. neous treatment is not feasible.
31. Radiofrequency Ablation of Osteoid Osteoma 395

If there is no safe access for a needle, treat-


ment is not possible.
Most importantly, vital structures within 1 cm
of the· tumor could be injured by heat. This is
especially true of nerves, making treatment of
spinal tumors problematic. A thin shell of bone
separating the lesion from the nerve may serve
as a relative barrier to thermal transmission
(50); however, each spinal case must be consid-
ered on its own merits, and we regard spinal
location as a relative contraindication.
FIGURE 31.9. Reformatted CT images show a tumor
Proximity to a joint (Fig. 31.8) or a growth arising within the epiphyseal center of the greater
plate (Fig. 31.9) does not appear to represent trochanter adjacent to the open growth plate.
an important problem, as the radiofrequency
method is probably less destructive than either
the surgical alternative or the continued pres-
ence of the tumor in such a location. We are
unaware of any clinical consequences of treat- Preparation for Radiofrequency
ing a tumor adjacent to a joint or a growth Treatment
plate.
Most patients with osteoid osteoma are healthy
children or young adults, and a brief interview
will determine suitability for general anesthe-
sia. Preprocedure laboratory testing does not
seem necessary. Patients taking pain medica-
tion are told to continue until the night before
the procedure, despite the known prolongation
of bleeding time caused by these agents. Bleed-
ing has not been a problem, perhaps because
the radiofrequency treatment acts as a form of
cautery.
General anesthesia is desirable for most
cases. Osteoid osteomas are very sensitive to
touch, and pain caused by needle entry into the
tumor is much more intense than pain due to a
typical bone biopsy. It cannot be adequately
blocked by local anesthesia of the periosteal
tissue (27). Furthermore, the small size of the
tumors, the frequent presence of dense sur-
rounding bone, and the requirement that no
portion of the tumor be more than 5 mm from
the electrode make proper positioning of the
FIGURE 31.8. CT scan demonstrates an osteoid electrode both important and difficult. Any
osteoma arising in the subchondral bone of the
patient movement can significantly lengthen
acetabulum. Although treatment of a lesion in such
a location will probably produce some damage to the procedure. Although spinal anesthesia can
articular cartilage, radiofrequency ablation is less often produce adequate analgesia, it is not suit-
destructive than any other intervention and proba- able for children, and often requires a longer
bly less damaging to the joint than prolonged pres- interval of recovery before the patient can be
ence of an untreated tumor. discharged.
396 D.I. Rosenthal and H. Ouellette

Once anesthesia is established, a localizing


scan using collimation of 3mm or less is per-
formed. This preliminary scan is used to map
the dimensions of the tumor and to establish
the best access. Several points should be borne
in mind in planning the procedure:

1. No part of the tumor must be more than


5 mm from the exposed tip of the electrode. For
lesions that measure more than 1 cm in length,
at least two electrode placements are required.
Similarly, smaller lesions may require multiple
placements if the electrode is not centrally
located on the first placement.
2. If it is necessary to penetrate dense cor-
tical or periosteal bone, it is preferable to FIGURE 31.10. Direct access to this lesion on the
approach the surface with the drill or needle posterior surface of the femoral neck was deemed
perpendicular to the surface to minimize the inadvisable because of the proximity to the sciatic
risk of skidding along the cortex. nerve, and the increased difficulty of anesthesia in
3. In most cases, the shortest path through the prone position. An anterior approach was used,
the bone is preferred. However, in some which required drilling through the entire femoral
instances it may be necessary or advisable to neck.
select an approach that requires more extensive
drilling. It is perfectly feasible to access a lesion
from the opposite side of the bone (Fig. 31.10); spinal needle) and scans are performed to
however, such an approach requires very confirm the level prior to making an incision.
careful attention (in all three dimensions). An
error made during the early stages of drilling is
not easily corrected once the needle is embed- Equipment
ded in bone.
4. Thermal diffusion will kill tissue around We have found the following combination of
the tip of the electrode for a distance of 5 to equipment to work well under a great variety
6 mm. For superficial lesions, there is a risk of of circumstances. Access to the bone is obtained
skin burn. Although cortical bone appears to using a Bonopty penetration set (Bonopty
confer a margin of safety (41,50), anatomic Bard, Radi Medical Systems, Uppsala,
boundaries are not necessarily thermal bound- Sweden). This is a disposable hand-operated
aries and therefore treatment may not be advis- drill that is placed with a protective cannula. It
able even though needle placement may be comes in one size only. It readily penetrates
accomplished safely. We generally decline to periosteal bone, trabecular bone, and thin cor-
perform RFA if adequate treatment of a lesion tices. It also can be advanced through solid
requires electrode placement less than 1cm cortical bone (such as might be found in the
away from the spinal cord or an important midfemur of a young adult) with considerable
nerve. effort and frequent interruptions to clear the
drill channels. When the drill reaches to within
For all imaging after the initial localizing 1 mm or less of the tumor, it is exchanged for a
scans, tube current often can be reduced to half I6-gauge, I5-cm Osty-Cut bone biopsy needle
or even less than that used for routine imaging (Bard!Angiomed, GmbH & Company Medi-
to minimize radiation exposure (Fig. 31.11). zintechnik KG) (Fig. 31.12).
Sterile skin preparation is followed by place- When the biopsy device enters the tumor, the
ment of a marker needle (usually a 22-gauge patient often exhibits sudden increases in res-
31. Radiofrequency Ablation of Osteoid Osteoma 397

The biopsy needle is withdrawn, leaving the


coaxial drill cannula in place, and the biopsy
specimen is sent for routine processing. We
have not found frozen section analysis to be
advantageous because frozen preparation is
suboptimal. The radiofrequency electrode is
introduced through the cannula into the hole
created by the biopsy needle.
Various electrodes are available on the
market. Most of these have been designed for
treatment of much larger soft tissue lesions and
are not optimal for osteoid osteoma. For the
greatest safety, the active tip of the electrode
should not exceed the dimensions of the tumor.
An electrode that results in a sphere of tissue
necrosis approximately 1 to 1.5 cm in diameter
A is ideal.
The dimensions of the electrode must be
appropriate to the type of needle that was used
for access. The electrode must be of small
enough caliber and great enough length to fit
through the device used to gain access to the
bone, and into the hole made by the biopsy
needle. We use a monopolar electrode with an
outer diameter (OD) of 1.1 mm and an exposed
tip of either 5- or 8-mm length (Radionics,
Burlington, MA). The smaller electrode is used
for average lesions up to approximately 7 or
8 mm. The larger electrode is used for bigger
tumors.
Check scans are performed following elec-
trode placement. It is important to confirm that

B
FIGURE 31.11. (A) An image from the initiallocaliz-
ing series performed using conventional exposure
factors (140kVp, 170mA). (B) Repeated scans used
for needle positioning are done with lower technical
factors (120kVp, 70mA in this case) to reduce radi-
ation exposure. Although there is increased mottle
on the images, the quality is adequate to perform the
procedure.

piratory rate and heart rate (27). This response


helps to identify needle placement within the
FIGURE 31.12. The top three devices are components
tumor, but placement also should be confirmed of a Bonopty penetration set, consisting of a cannula,
by check scans. The biopsy needle is passed stylette, and drill. The lower two are the Osty-Cut
through the entire lesion. Plugging the needle bone biopsy needle and its stylette. Notice that the
with bone on the far side of the lesion helps to Osty-Cut device is long enough to obtain a biopsy
retain the specimen upon needle withdrawal. through the Bonopty cannula.
398 D.1. Rosenthal and H. Ouellette

no portion of the tumor is more than 5 or 6 mm Steri-Strip is adequate. No suture, cast, or brace
away from the exposed portion of the is required.
electrode. This is easy to confirm for in-plane Upon awakening from anesthesia, the
distances; however, longitudinal extent may patient often, but not invariably, complains of
require review of several images. If a part of the pain. The pain is similar to tumor pain in char-
tumor is farther away, an additional drill hole, acter and location, but may be more intense. It
biopsy, and treatment should be performed is characteristically most severe within the first
to encompass the portion of the tumor not hour, and once brought under control with nar-
covered by the initial thermal lesion. Curiously, cotics it steadily improves. It may be necessary
the performance of a radiofrequency treatment to send the patient home with a prescription for
has no short-term effect upon the inter- a narcotic medication. One or two doses should
pretability of the biopsy, and the pathologist be adequate, after which the patient may switch
will not be able to identify the fact that the to nonsteroidals, or may not require medica-
second specimen has been heated. tion. Approximately half of the patients are
Using manual or automatic output controls able to recognize that the tumor pain is gone by
on the generator, the temperature at the tip of the morning following the procedure, although
the electrode is gradually (approximately 1 there may be residual procedure-related pain
degree/second) increased until it reaches 90°C. for a few days.
Downward adjustment of the generator output Patients are allowed to resume daily activi-
is required to maintain this temperature as pro- ties immediately. For tumors located in weight-
gressive coagulation of the lesion occurs. The bearing cortex, such as the upper femur,
exact time and temperature for treatment is dif- patients are advised to avoid activities in-
ficult to rationalize on purely scientific grounds. volving distance running for 3 months follow-
The ultimate size of the thermal lesion depends ing the procedure, even though we have not
on the opposing effects of thermal diffusion, observed any postprocedure stress fractures.
and tissue cooling, largely due to blood flow. In Leg muscle atrophy and abnormal gait (if
the steady state, temperature falls as the fourth present) are expected to resolve spontaneously.
power of the distance from the electrode (38), We have not required assistance from physical
leading to a rather abrupt margin between therapists.
viable and destroyed tissue. Since the steady
state is reached shortly after the target tem- Follow-Up
perature is achieved, sustained application of
energy would appear to have little purpose. Our routine is to see the patient in the recovery
However, our experience with 4 minutes of room to decide on postprocedure medication
heating seemed to result in more recurrences needs, and to speak by telephone the next
than a 6-minute treatment, which is our current morning to answer questions. One week later
standard. Imaging after treatment is not we again telephone to report the results of the
required and provides no useful information. biopsy, and to assess the short-term outcome.
All subsequent follow-up is performed by the
Care After the Procedure referring clinician.
We do not believe that postprocedure
Approximately 10 to 20 minutes before com- imaging is essential in asymptomatic patents.
pletion, a nonsteroidal antiinflammatory med- However, if performed, at approximately 6
ication (Ketorolac, Allergan Pharmaceuticals, weeks after the procedure, it frequently reveals
30mg IV adult dose) is administered intra- a spherical radiolucent "halo" approximately 1
venously to help with postprocedure pain. If the to 1.5 cm from the electrode site, presumably
lesion is superficially located on the bone, injec- representing the margins of the necrotic area.
tion of local long-acting anesthetic also may be Subsequently the periosteal bone produced
helpful. However, in many instances it does not by the lesion becomes incorporated into the
appear to be effective. A simple bandage or normal cortex and the tumor becomes increas-
31. Radiofrequency Ablation of Osteoid Osteoma 399

ingly and uniformly ossified. Young children osteoid osteoma. Clin Orthop 2000;373:115-
may have completely normal imaging studies 124.
6 months after the procedure. Older individu- 2. de Berg JC, Pattynama PM, Obermann WR,
als may still demonstrate focal sclerotic areas Bode PJ, Vielvoye GJ, Taminiau AH. Percuta-
neous computed-tomography-guided thermoco-
even after several years. If the patient has no
agulation for osteoid osteoma. Lancet 1995;
symptoms, the appearance is irrelevant.
346(8971):350-351. (Notes: 18 patients with no
complications using the ablation method. Patient
hospitalized overnight and discharged next
morning. Resumed normal activities immedi-
Success Rates ately. All remained pain free except one; flu 3-15
months. Second treatment eliminated symptoms
Success of treatment is judged by freedom in the unsuccessful case.)
from pain and tumor recurrence. These are not 3. Lindner NJ, Ozaki T, Roedl R, Gosheger G,
necessarily identical, as patients may have Winkelmann W, WortIer K. Percutaneous radio-
persistent pain for various reasons other than frequency ablation in osteoid osteoma. 2001;
recurrence. 83B(3):391-396. (Notes: Westfalishe Wilhelms
In our experience it is extremely infrequent Universitat, Munster, Germany.)
for a patient to obtain no relief at all from 4. Pinto CH,TaminiauAHM,Vanderschueren GM,
Hogendoorn PCW, Bloem JL, Obermann WR.
treatment. However, if there is residual
Technical considerations in CT-guided radiofre-
tumor, recurrence of symptoms usually follows
quency thermal ablation of osteoid osteoma:
after a relatively brief interval. On average, tricks of the trade. AJR Roentgenol 2002;
recurrence is apparent between 3 and 6 months 179(12):1633-1642.
after treatment. We have seen occasional cases 5. Rosenthal DI, Hornicek FJ, Wolfe MW, Jennings
as late as 18 months. Although the literature CL, Gebhardt MC, Mankin HI Percutaneous
documents rare instances after many years radiofrequency coagulation of osteoid osteoma
(51,52), most authorities agree that a 2-year compared with operative treatment. J Bone
pain-free interval is sufficient to document Joint Surg 1998;80A(6):815-821.
cure. 6. Mirra I Bone Tumors. Philadelphia: JB Lippin-
Using this criterion, in our experience cott, 1980:97.
7. Gitelis S, Schajowicz F. Osteoid osteoma and
approximately 90% of patients have complete
osteoblastoma. Orthop Clin North Am 1989;
and permanent relief of symptoms following a 20(3):313-325.
single treatment, 5% have recurrences, and 5% 8. Greenspan A. Benign bone-forming lesions:
have some form of persistent symptoms but do osteoma, osteoid osteoma, and osteoblastoma.
not desire any further evaluation or therapy Skel Radiol 1993;22(7):485-500.
(16). Other series document somewhat higher 9. Habermann ET, Stern RE. Osteoid-osteoma of
rates of recurrence (53). Recurrence rates fol- the tibia in an eight-month-old boy. J Bone Joint
lowing operative intervention appear to be Surg Am 1974;56:633-636.
similar (51). 10. Szabo RM, Smith B. Possible congenital-osteoid
Recurrences are almost invariably in the osteoma of a phalanx. J Bone Joint Surg Am
same location as the initial tumor, or immedi- 1985;67:815-816.
11. Greco F, Tamburrelli F, Laudati A, La Cara A,
ately contiguous, and present with the same
Trapani G. Nerve fibres in osteoid osteoma. Ital
symptoms. The treatment options for a recur- J Orthop TraumatoI1988;14(1):91-94.
rent lesion are identical to that for the initial 12. Wold LE, Pritchard DJ, Bergert J, Wilson DM.
lesion (medical, surgical, or percutaneous), Prostaglandin synthesis by osteoid osteoma
although the success rates are lower. and osteoblastom. Mod Pathol 1988;1(2):129-
130.
13. Esquerdo J, Fernandez CF, Gomar F. Pain in
osteoid osteoma: histological facts. Acta Orthop
References Scand 1976;47(5):520-524.
1. Barei DP, Moreau G, Scarborough MT, Neel 14. O'Connell JX, Nanthakumar SS, Nielson GP,
MD. Percutaneous radiofrequency ablation of Rosenberg AE. Osteoid osteoma: the uniquely
400 D.L Rosenthal and H. Ouellette

innervated bone tumor. Mod PathoI1998;11(2): 29. Hasegawa T, et al. Intracortical osteoblastic
175-180. osteosarcoma with oncogenic rickets. Skel
15. Kiers L, Shield LK, Cole WG. Neurological man- RadioI1999;28:41-45.
ifestations of osteoid osteoma. Arch Dis Child 30. Abril lC, Castillo F, Diaz A. Brodie's abscess of
1990;65(8):851-855. the hip simulating osteoid osteoma. Orthopedics
16. Chiou Y-Y, Rosenthal DI, Rosenberg AE. 2000;23(3):285-287.
"Beaded" osteoid osteoma: a possible interme- 31. Golding JSR. The natural history of osteoid
diate between solitary and multicentric tumor. osteoma. J Bone Joint Surg 1954;36B(2):218-229.
Skel RadioI2003;32(7):412-415. 32. Kneisl JS, Simon MA. Medical management
17. Kenan S, Abdelwahab IF, Klein Ml, Hermann G, compared with operative treatment for osteoid
Lewis MM. Case report 864. Elliptical, multicen- osteoma. 1 Bone Joint Surg 1992;74A(2):179-
tric periosteal osteoid osteoma. Skel Radiology 183.
1994;23(7):565-568. 33. Leicester AW, Trantalis IN. Osteoid osteoma in
18. Keret D, Harcke HT, MacEwen GD, Bowen lR. a young child: successful non-operative manage-
Multiple osteoid osteomas of the fifth lumbar ment. ANZ 1 Surg 2001;71:491-493.
vertebra. Clin Orthop Rei Res 1989;248:163- 34. Saville Po. A medical option for the treatment
168. of osteoid osteoma. Arthritis Rheum 1980;23:
19. O'Dell CW lr, Resnick D, Niwayama G, George 1409-1411.
TG, Linovitz RJ. Osteoid osteoma arising in 35. Jackson RP, Reckling FW, Mantz FA. Osteoid
adjacent bones. Report of a case. 1 Canad Assoc osteoma and osteoblastoma. Clin Orthop Rei
Radiol 1976;27:298-300. Res 1977;128:303-313.
20. Rand JA, Sim FH, Unni KK. Two osteoid osteo- 36. Kayser M, Muhr G. Eighteen-year anamnesis of
mas in one patient: a case report. J Bone Joint osteoid osteoma-a diagnostic problem? Arch
Surg 1982;64A(8):1243. Orthop Trauma Surg 1988;107(1):27-30.
21. Schai F, Friederich N, Kruger A, Jundt G, Herbe 37. Vickers CW, Pugh DC, Ivins Je. Osteoid
E, Buess P. Discrete synchronous multifocal osteoma: a fifteen-year follow-up of an un-
osteoid osteoma of the humerus. Skel Radiol treated patient. J Bone Joint Surg Am 1959;
1996;25(7):667-670. 41A(2):357-358.
22. Alani WO, Bartal E. Osteoid osteoma of the 38. Antich PP, Pak CYC, Gonzales J, Anderson 1,
femoral neck stimulating an inflammatory Sakhaee K, Rubin e. Measurement of intrinsic
synovitis. Clin Orthop Rei Res 1987;223:308- bone quality in vivo by reflection ultrasound:
312. correction of impaired quality with slow-release
23. Davies M, Cassar-Pullicino VN, Davies AM, sodium fluoride and calcium citrate. J Bone
McCall IW, Tyrrell PN. The diagnostic accuracy Miner Res 1993;8(3):301-311.
of MR imaging in osteoid osteoma. Skeletal 39. Foeldvari I, Schmitz Me. Rapid development of
Radiol 2002;31(10):559-569. severe osteoarthritis associated with osteoid
24. Abdelwahab IF, Hermann G, Lewis MM, Klein osteoma in a young girl. Clin Rheumatol 1998;
MJ. Case report 588: intracortical chondroma 17(6):534-537.
of the left femur. Skel Radiol 1990;19:59-61. 40. Weber KL, Morrey BE Osteoid osteoma of the
25. Rudman DP, Damron TA, Vermont A, Mathur S. elbow: a diagnostic challenge. 1 Bone Joint Surg
Intracortical chondroma. Skel Radiol 1998; Am 1999;81A(8):1111-1119.
27(10):581-583. 41. Cove lA, Taminiau AH, Obermann WR,
26. Bauer TW, Dorfman HD, Latham IT. Periosteal Vanderschueren GM. Osteoid osteoma of the
chondroma: a clinicopathologic study of 23 cases. spine treated with percutaneous computed
Am 1 Surg PathoI1982;6(7):631-637. tomography-guided thermocoagulation. Spine
27. Ramnath RR, Rosenthal DI, Cates J, Gebhardt 2000;25(10):1283-1286.
M, Quinn RH. Intracortical chondroma simulat- 42. Gangi A, Dietemann JL, Gasser B, et al.
ing osteoid osteoma treated by radiofrequency. Interventional radiology with laser in bone
Skeletal Radiol 2002;31:597-602. and joint. Radiol Clin NorthAm 1998;36(3):547-
28. Erickson JK, Rosenthal DI, Zaleske DJ, Geb- 557.
hardt MC, Cates 1M. Primary treatment of chon- 43. Witt ID, Hall-Craggs MA, Ripley P, Cobb IP,
droblastoma with percutaneous radio-frequency Bown SG. Interstitial laser photocoagulation for
heat ablation: report of three cases. Radiology the treatment of osteoid osteoma. J Bone Joint
2001;221(2):463-468. Surg 2000;82B(8):1125-1128.
31. Radiofrequency Ablation of Osteoid Osteoma 401

44. Amendola A, Vellet D, Willits K. Osteoid 49. Sanhaji L, Gharbaoul IS, Hassani RE, Chakir N,
osteoma of the neck of the talus: percutaneous, Jiddane M, Boukhrissi N. A new treatment of
computed tomography-guided technique for osteoid osteoma: percutaneous sclerosis with
complete excision. Foot Ankle Int 1994;15(8): ethanol under scanner guidance. J Radiol 1996;
429-431. 77(1):37-40. (Original in French.)
45. Assoun J, Railhac J-J, Bonnevialle P, et al. 50. Dupuy DE, Hong R, Oliver B, Goldberg SN.
Osteoid osteoma: percutaneous resection with Radiofrequency ablation of spinal tumors: tem-
CT guidance. Radiology1993;188(2):541-547. perature distribution in the spinal canal. AJR
46. Atar D, Lehman WB, Grant AD. Tips of the 2000;175(5):1263-1266.
trade. Computerized tomography-guided exci- 51. Norman A. Persistence or recurrence of pain: a
sion of osteoid osteoma. Orthop Rev 1992; sign of surgical failure in osteoid osteoma. Clin
21(12):1457-1458. Orthop ReI Res 1978;130:263-266.
47. Sans N, Galy-Fourcade D,Assoun J, et al. Osteoid 52. Regan MW, Galey JP, Oakeshott RD. Recurrent
osteoma: CT-guided percutaneous resection and osteoid osteoma. Clin Orthop ReI Res 1990;253:
follow-up in 38 patients. Radiology 1999;212(3): 221-224.
687-692. 53. Vanderschueren GM, Taminiau AHM, Ober-
48. Duda SH, Schnatterbeck P, Harer T, Giehl J, mann WR, Bloem JL. Osteoid osteoma: clinical
Bohm P, Claussen CD. Treatment of osteoid results with thermocoagulation. Radiology 2002;
osteoma with CT-guided drilling and ethanol 224(1):82-86.
instillation. Dtsch Med Wochenschr 1997;
122(16):507-510.
32
Image-Guided Prostate Cryotherapy
Gary Onik

Background of Cryotherapy 229 patients followed for up to 10 years. Com-


parison with radical prostatectomy and radia-
The treatment of prostate cancer remains a sig- tion therapy patients showed equal survival
nificant dilemma. The prevalence of prostate among the various treatment modalities.
cancer is quite high, documented by pathologic Although cryosurgery showed some advan-
studies. It is well accepted that a major propor- tages, such as being able to treat patients with
tion of prostate cancers will not be clinically sig- large bulky tumors, poor monitoring of the
nificant (1). Even those cancers with a volume freezing process resulted in complications such
of 0.5 cc or greater have a variable biologic as urethrocutaneous and urethrorectal fistulas
behavior. On the other hand, the treatments for that limited acceptance of the procedure.
prostate cancer are not without substantial In 1993 Onik et al (5) reported the first
chance for lifestyle-limiting morbidity. Recent series of percutaneous ultrasound-guided and
information showing minimal survival benefit -monitored prostate cryotherapy (Figs. 32.1
between no treatment and radical prostatec- and 32.2). This report resulted in a resurgence
tomy has emphasized the concept of "watchful of interest. As with any new procedure, ultra-
waiting"; that is, not treating the primary tumor sound-guided prostate cryotherapy went
at all has gained acceptance as a viable manage- through a learning curve. Reported results
ment alternative in certain patient populations about the control of the cancer as well as com-
(2). The decision to treat an individual's prostate plications were variable. A negative perception
cancer with a particular therapy or at all, of the procedure was compounded in that most
requires a careful assessment of the risk versus early series predominantly treated patients who
benefit for that patient. As the complications had failed radiation therapy and who had much
and lifestyle limiting side effects of treatments higher complication rates, particularly that of
are reduced, these decisions become easier. The incontinence (73%) (6,7). The situation was
reintroduction of ultrasound-guided prostate also exacerbated by a high urethral complica-
cryotherapy is founded on the goal of decreas- tion rate, caused by use of a rigged urethral
ing the morbidity of prostate cancer treatment. warming catheter, when the original catheter
Gondor et al (3) in 1966 first reported the was placed back into investigational use by the
concept of a cryotherapy procedure for the Food and Drug Administration (FDA) (14% vs.
treatment of prostate disease. Subsequently, an 42 %) (8). Despite these early problems, the
open transperineal cryosurgery procedure was long-term results from multiple institutions
developed in which the freezing was carried were examined, and finally the Health Care
out on the surface of the prostate with visual Financing Administration (HCFA) in 1999
monitoring. Using this same approach, Bonney removed cryosurgery from the investigational
et al (4) reported results of this procedure in category, placing it alongside radiation and

402
32. Image-Guided Prostate Cryotherapy 403

chemical disease-free survival rate with


cryotherapy for the low-risk group was 76%,
and for comparable brachytherapy the
reported series was 75% to 87% and for con-
formal radiotherapy 67% to 81 %. The results
were similarly comparable for medium- and
high-risk groups. In terms of complications, the
incidence of rectal complications was greater
for radiation therapy, while the rate of impo-
tence was greater for cryotherapy. Long et al
noted that it was remarkable that the results
were so similar, despite the substantial dispar-
ity in resources and experience between the
FIGURE 32.1. The transperineal placement of cryo- two therapies. As we will examine, when the
probes into the prostate using transrectal ultrasound latest advances in cryotechniques are consid-
(US) guidance. This is the same approach used for
ered, the results and complication rates for
brachytherapy.
cryosurgery reported by Long et al can be
markedly improved.
radical prostatectomy as a treatment for
primary prostate cancer. Patient Selection
The quality of these results, taking into
account the early technical variability of the When dealing with prostate cancer therapy, the
procedure, as well as the urethral warmer extent and pathologic character of the patient's
problems, is best reflected in the multiinstitu- disease are essential in choosing the proper
tional study recently published by Long et al therapy. Treatments such as radical prostatec-
(9). When they compared cryotherapy to radio- tomy and radiation have higher recurrence
therapy using the same patient risk stratifica- rates as the extent and aggressiveness (Gleason
tion and success criteria, the results were score) of the patient's cancer increase. An
comparable to both external-beam radiation advantage of cryotherapy is the flexibility of the
therapy and brachytherapy. The 5-year bio- procedure to be tailored to treat both high- and
low-risk patients, as well as patients who have
failed radiation therapy.

The Patient at High Risk for


Local Recurrence
The use of cryotherapy for the treatment of
solid organ cancers made a resurgence with the
advent of ultrasound monitoring of hepatic
cryosurgery in 1984 (10). Hepatic cryotherapy
filled a unique place in the armamentarium of
liver cancer treatment, in that it successfully
treated patients with multiple tumors or tumors
that were unresectable due to proximity to
FIGURE 32.2. Transrectal ultrasound showing excel- major vasculature that could not be sacrificed
lent visualization of the freezing front (FF) as a (11,12). Due to its target patient population
hyperechoic rim with posterior acoustic shadowing. of previously untreatable patients, with an
R, rectal wall; SV, seminal vesicle; UG, urogenital expected mortality of virtually 100%, imaging-
diaphragm. guided hepatic cryotherapy was readily
404 G.Onik

embraced by the surgical oncologic community analysis, is more problematic, since the exact
(13). margin status of the tumor is not known
In at least one way, prostate cancer is akin to preoperatively or postoperatively following
liver cancer in that many patients with prostate cryotherapy.
cancer are unresectable, based on the likeli- One study, in which aggressive periprostatic
hood of capsular penetration at the time of freezing was facilitated by separating the
definitive treatment. Unfortunately, efforts at rectum from the prostate at the time of the
preoperative staging have been inadequate to operation by a saline injection into Denonvil-
identify this patient population. The difficulty liers' fascia, showed no local recurrences
of the situation is further compounded because demonstrated in 61 patients followed for up to
at the time of radical prostatectomy the 4 years (20). These good results occurred
surgeon's ability to appreciate capsular pene- despite the fact that 68% of the patients were
tration and involvement of the neurovascular considered at high risk of capsular penetration
bundles is inadequate. In a recent study by and local failure, based on a Gleason score of 7
Vaidya et al (14), there was virtually no corre- or greater, PSA more than lOng/mL, having
lation between the surgeon's determination of failed radiation therapy, or extensive bilateral
tumor penetration into the periprostatic tissue disease based on preoperative biopsies.
with involvement of the neurovascular bundle When cryoablation is carried out aggres-
and actual pathologic confirmation. Thus, posi- sively with the aim of a negligible PSA
tive margin rates of 30% to 40% associated post-treatment, comparison with radical prosta-
with nerve sparing radical prostatectomy are tectomy should be possible. In the only pub-
the usual situation. lished study comparing the outcomes between
Using various clinical parameters such as the two (21), cryotherapy had a 23% greater
Gleason score, clinical stage, and prostate- chance of resulting in a PSA of less than
specific antigen (PSA) level, preoperative O.2ng/mL than did radical prostatectomy (96%
prediction of the statistical chance for capsular vs. 73 %). When a 0 PSA was used as the success
penetration can be reasonably assessed (15,16). criterion, cryotherapy maintained an approxi-
This approach can lower the positive margin mately 20% advantage over radical prostatec-
rate when rigorously applied, as demonstrated tomy (66.9% vs. 48.2%). As risk for positive
by Eggleston and Walsh (17). In clinical prac- margins increased, based on a PSA of 20 or
tice, however, most urologists believe that greater, cryotherapy maintained these results,
radical prostatectomy still is the gold standard while those of radical prostatectomy deterio-
of treatment. This leads to a natural reluctance rated further (86% vs. 36%). While this study
to deny a patient the treatment they feel is was retrospective and with only a relatively
unproven for cure. Based on the success of small number of patients, the results are con-
cryotherapy in treating unresectable liver sistent with the original rationale of destroying
cancer, it was hoped that with the ability to extracapsular cancer by treating the peripros-
freeze into the periprostatic tissue, thereby tatic tissues. In addition, these findings are
encompassing tumor capsular penetration, that consistent with other studies that show success
prostate cryotherapy could improve the in treating T3 disease and other high-risk
outcome in those patients at high risk of posi- patients.
tive surgical margins. In two recent cryotherapy reports with 5- and
Numerous studies on ultrasound-guided 7-year follow-up in medium- and high-risk
prostate cryotherapy have demonstrated suc- patients, the results were superior to other
cessful treatment of patients with stage T3 treatment modalities, including conformal
prostate cancer with demonstrated gross extra- radiation therapy, brachytherapy, and radical
capsular disease (18,19). Demonstration of this prostatectomy (22,23). Kaplan-Meyer curves
concept in patients with a "high likelihood" of showed virtually no difference among low-,
capsular penetration, based only on statistical medium-, and high-risk patients. These results
32. Image-Guided Prostate Cryotherapy 405

are remarkable, and have major implications garnering patients who are interested in main-
for overall survival in patients who previously taining sexual function. In our experience, total
had a poor prognosis. These results, coupled cryotherapy has the same impotency rate as
with the low morbidity of the procedure, the non-nerve-sparing radical prostatectomy: vir-
ability to be performed in older patients and tually 100% in the short and intermediate
the ability to be repeated when needed, we terms. The same reason that cryotherapy has
believe make cryotherapy the procedure of demonstrated an advantage in treating high-
choice in this subset of patients. risk patients, however, contributes to this rate
of impotence. When cryoablation is properly
carried into the periprostatic tissue to encom-
pass any extracapsular tumor that preferen-
Organ-Confined, Low-Volume, tially invades the neurovascular bundles, both
Low Gleason Score Disease bundles are necessarily destroyed. Therefore, in
(Nerve-Sparing Cryotherapy) patients at high risk of bilateral extracapsular
When treating patients with prostate cancer disease, impotence is an expected side effect,
that is organ confined, low morbidity and and actually a welcome sign that proper treat-
limited effect on lifestyle become major factors ment has been carried out. The original thought
in selection of the treatment option. Cryother- that sexual dysfunction secondary to cryother-
apy has low morbidity, and the procedure can apy would only be temporary, based on the
be done on an outpatient basis. Also, there is no well-known ability of peripheral nerves to
need for blood transfusions and no reported regenerate after cryoablation, was erroneous.
incidence of periprocedural cardiovascular or The parasympathetic system, critical to erec-
pulmonary complications. The most dreaded tion, has its secondary cell bodies in proximity
local complication of prostate cryoablation, to the prostatic capsule; the destruction of these
urethrorectal fistula, which occurs due to rectal by cryotherapy is irreversible.
freezing, has virtually been eliminated by the However, nothing limits cryotherapy from
technique of injecting saline into Denonvilliers' being carried out as a nerve-sparing procedure,
fascia at the time of the procedure, thus creat- similar to the approach with radical prostatec-
ing a large buffer zone between the rectum and tomy. The ability of cryotherapy to be repeated
the prostate capsule. When failed radiation is an advantage. Temperature monitoring to
cases and those patients treated without the prevent freezing of the contralateral neuro-
original urethral warmer are excluded, vascular bundle from the tumor can be used to
cryotherapy has an excellent record in relation preserve this structure, and therefore maintain
to urinary complications, with low incontinence potency.
rates of 2 % or less (20,24,25). A study pub- We have followed a group of nine patients, for
lished by Robinson et al (26), looking at the as long as 6 years, who underwent unilateral
quality of life of patients undergoing cryoabla- nerve-sparing cryotherapy. The indication for
tion, showed return of urinary function to base- nerve-sparing cryotherapy was the small
line status in virtually all patients by 1 year volume of cancer demonstrated on biopsy,
postprocedure. along with the demonstration of unilateral
In an effort to prove itself as a viable cancer disease by staging procedures. The potency rate
treatment, cryotherapy has been aggressively (defined as erection sufficient to carry out inter-
aimed at total prostate gland ablation. It is course to the satisfaction of the patient) was
clear, however, that total gland ablation with 78% (seven of nine patients), with no patient
freezing of both neurovascular bundles has a having evidence for a local recurrence on PSA
significant negative impact on sexual function monitoring or biopsy. The group included high-
(26). Cryotherapy's high rate of impotence is a risk patients, with five of nine having some risk
major disadvantage compared to nerve-sparing factor for extracapsular disease (three of nine
radical prostatectomy and brachytherapy in patients had a Gleason score of 7 or above). The
406 G. Onik

potency results were surprising in that only one The treatment of patients for salvage after
nerve bundle was spared, but this lack of manip- radiation therapy was not without difficulties
ulation spares injury to the blood supply. These however. Early findings showed poor cancer
results rival bilateral nerve sparing of radical control results with fewer than 25% of patients
prostatectomy (27). The high potency rate with reaching PSA levels of 0.2 ng/mL (8) or less, and
sparing of only one neurovascular bundle is with positive biopsy rates as high as 35%. Com-
better than that expected with radical prostatec- plications in this patient population also were
tomy, which has been reported as 13% to 41 % significant. While the incontinence rate for non-
(28,29). Brachytherapy has a reported potency salvage cryotherapy patients is less than 2%,
rate of 49% 2 years postprocedure (30). significant incontinence for radiation salvage
Nerve-sparing cryotherapy is a blend of an patients can be as high as 46% (33). Advances
aggressive, yet minimally invasive procedure, in cryotherapy technique have improved these
which accounts for the combination of excel- results; a study by Ghafar et al (34) demon-
lent cancer control and lack of complications. strated a 60% success rate at providing an
Extensive freezing of the periprostatic tissue is undetectable PSA; the associated incontinence
still carried out on the side of the demonstrated rate was 9%.
tumor. In patients with a high Gleason score, or Our results are consistent with this afore-
with cancer demonstrated at the base of the mentioned report. Twenty-four patients who
gland, prophylactic freezing of the confluence had locally recurrent prostate cancer after
of the seminal vesicles also can be carried radiation therapy were treated with cryother-
out. The expected incidence of urinary and apy with a mean follow-up of 3 years. The group
rectal complications is lower than that for had a high risk of recurrence-15 of 24 patients
total cryoablation, radical prostatectomy, or had a Gleason score of 7 or above, and four
brachytherapy. No patient in our series demon- patients had already failed hormone therapy.
strated persistent incontinence. Importantly, The median preoperative PSA was 4.7ng/mL,
any errors in patient selection are correctable standard deviation (SD) 3.4ng/mL, and the
by re-treatment without added morbidity. We postcryotherapy median PSA was 0.01 ng/mL
believe this treatment approach has major with a SD of 1.4. Of the 24 patients, 19 (79%)
implications, and, if confirmed, could become have a stable PSA that is undetectable. Of the
the treatment of choice for patients with low- five unstable patients, all have had negative
risk disease. biopsies. Thus, in these 24 high-risk patients,
there is no evidence of local recurrence. In
addition, three of the four patients who had
Patients with Local Recurrence After failed hormonal therapy have no evidence of
Radiation Therapy
recurrent disease. The incontinence rate in
Patients who have received a maximal dose of these patients was 33%, but does not reflect
radiation but suffer local recurrence of tumor the recent changes in freezing protocols and
without evidence of metastatic disease, theo- urethral warmer changes that have lowered this
retically are still curable. Unfortunately, radia- rate to 10% (34). In addition, patients with
tion destroys the tissue planes needed for a safe severe incontinence that limited lifestyle were
and effective attempt at salvage radical prosta- treated successfully with artificial sphincter
tectomy. Radical prostatectomy in a salvage placement.
situation has reported positive margins in 40% Based on these results, cryotherapy recently
of patients (31) with prohibitive morbidity, has been approved by Medicare as the only
demonstrating a 58% incontinence rate and an treatment specifically for the indication of
incidence of rectal injury as high as 15% (32). recurrent cancer after radiation failure.
Consequently, salvage radical prostatectomy Based on the potentially curative nature of
rarely is performed in this setting; most patients the procedure, cryotherapy has become the
are placed on palliative hormonal ablation procedure of choice in this difficult group of
therapy. patients.
32. Image-Guided Prostate Cryotherapy 407

Technical Advances in
Cryotherapy
Saline Injection into
Denonvilliers' Fascia
A major theoretical criticism of prostate
cryoablation has been based on the anatomy of
the pelvis, in particular the proximity of the
prostate capsule to the rectal mucosa. Limited
space between the prostate and rectum can robe A
result in freezing of the rectal mucosa with
resultant urethrorectal fistula. Conversely, fear
of urethrorectal fistula can lead to premature
cessation of the freezing process. This can lead
to a high incidence of tumor recurrence. Even
for the experienced operator, it can be said
that the procedure at times is a nerve-wracking
balancing act between adequate treatment and
rectal injury.
Probably the most important advance in the
technique of prostate cryoablation that leads to B
favorable reproducible results relates to the
FIGURE 32.3. (A) The placement of a needle into
injection of saline into Denonvilliers' fascia at Denonvilliers' fascia between the rectum (R) and
the time of freezing to temporarily increase the the prostate capsule. There is a urethral warming
space between the rectum and prostate (Fig. catheter in place. (B) This diagram shows an increase
32.3). In over 200 cases, this maneuver has in the space between the rectum and the prostate
virtually eliminated the risk of rectal freezing after saline has been injected.
and the complication of urethrorectal fistula
without adding any additional morbidity in our
experience. the posterior urethral region to encompass the
The elimination of the fear of rectal freezing confluence of the seminal vesicles, thereby pre-
has numerous consequences that improve the venting recurrence in this region. Last, with this
results of cryotherapy. First, with the rectum out move of cryoprobes into the peripheral zone,
of the way, freezing can be extended "Outside the freezing of the periurethral tissue, while still
the prostate sufficiently to bring the -35°C adequate, is less intense, thereby decreasing
isotherm to the capsule of the prostate; this the possibility of urethral sloughing. With adop-
ensures adequate temperatures for cancer tion of this technique, the learning curve is
destruction throughout the prostate. Second, shortened, and the future reproducibility of
freezing can be extended far enough outside cryotherapy results is assured.
the prostate to include extracapsular extension
of cancer; this improves local control of cancer
in high-risk patients. In addition, with more
Temperature Monitoring in
freezing room, cryoprobes can be placed
Critical Areas and Improved
farther into the peripheral zone of the gland,
Cryoablation Protocols
where 80% of cancers reside. This exposes For reliable destruction of cancer by freezing,
these cancers to faster freezing and colder temperatures must reach certain critical limits.
temperatures, both of which improve cancer Recent in vitro and in vivo studies have shown
destruction. The arrangement of the cryoprobes that reaching -35°C with at least two freeze-
also can change with a probe being placed into thaw cycles is needed to destroy prostate cancer
408 G. Onik

cells reliably (35). Marked improvement in Increasing the number of probes beyond
clinical results occurs when temperature is eight could have a potentially negative effect.
monitored by thermocouples placed in critical Recent data generated by a "cryoseed system"
areas in the prostate and when two full from Galil Medical Inc. (Haifa, Israel) that
freeze-thaw cycles are performed. Wong et al utilizes 17-gauge needle probes to create a 1-
(7) had 10 failed cases in the first 12 with only cm-diameter iceball has shown a decreased
ultrasound monitoring; six failures occurred in ability to totally ablate the prostate gland.
the next 66 cases after thermocouple monitor- These degraded results probably are the result
ing was instituted. A major advantage of of the short freezing length of these probes and
cryotherapy, however, is the ability to re-treat difficulty in accurately overlapping the freezing
local failures. Using thermocouple monitoring zones along the length of the gland. All the data
and re-treating, the previous failed cases had a currently used to gain acceptance of cryoabla-
negative biopsy rate of 94% (72 of 77 patients) tion were developed with probes that freeze the
at 30 months. length of the gland in one freeze. Departing
Salikan et al (24) reported an almost identi- from this concept jeopardizes much of what has
cal experience in which 10 of 69 patients ini- been learned about how to obtain consistent
tially had positive biopsies after treatment, with results.
the majority of the 10 occurring early in their
cryotherapy experience. Ultimately, after re-
treatment, 68 of 69 patients (99%) had
Future Technical Improvements
negative biopsies. With recent approval for reimbursement by
Clearly, temperature monitoring in critical HCFA, there finally is an incentive for com-
areas of the prostate gland is essential to good mercial interests to make the investment to
cancer control using cryoablation. Temperature technically improve the procedure. At the
monitoring also may help limit complications present time the greatest efforts are being made
from overfreezing. Once the target temperature in improvements already well-established in
is reached, further freezing becomes a greater brachytherapy. Since the freezing capabilities of
liability. cryoprobes are highly predictable, planning
software to direct proper cryoprobe placement
based on gland size and shape is already
Improved Equipment with Increase being developed. Planning software shortly
from Five to Eight Probes will be coupled to guidance software and
The original LN2-based freezing equipment hardware that will greatly simplify what now
now has been replaced by Joule-Thompson is a totally freehand approach to cryoprobe
argon gas systems. These systems allow faster placement.
freezing rates that improve cancer destruction. While ultrasound is inexpensive and readily
The more precise control of the freezing available, it still is highly operator dependent
process by gas systems also adds to the safety and difficult to use by novices. Other cross-
of the procedure. sectional imaging techniques such as MRI have
Increasing the number of probes from five the potential to simplify and standardize
to eight has allowed a more uniform freezing cryotherapeutic procedures. As opposed to
temperature throughout the gland, which also ultrasound (US), in which ice causes shadowing
improves results. Lee et al (36) showed that and shows only the leading edge of the iceball,
there was a statistically significant difference in both MRI and CT demonstrate the full extent
total gland ablation (measured by PSA level of the freezing (Fig. 32.4). Using thermody-
and absence of glands on subsequent biopsy) namic equations, these images can be used to
between using five liquid nitrogen probes and noninvasively calculate temperatures within
six to eight argon gas probes (67% vs.89%). the iceball and automatically control the cryo-
Ultimately, Lee et al achieved a negative biopsy probes to create a specific freezing shape and
rate of 96% (157 of 163 patients). profile.
32. Image-Guided Prostate Cryotherapy 409

We believe, as the inherent advantages of


freezing as with brachytherapy radiation are
appreciated, further technical development will
continue to occur. Clearly, at the present time
we are seeing only the tip of the "iceberg" of
the technical potential of cryotherapy.

Conclusions
Ultimately, the choice of treatments also may
A involve outcomes and economic considerations.
This will require us to examine closely what the
overall costs are of providing the various treat-
ment options for prostate cancer. For instance,
how superior do the results of proton beam
therapy have to be over other treatment
options to justify building an $80 million
facility? Can the expense of providing
brachytherapy and external beam radiation in
combination be justified if the same patient
population can be treated with equally good
results by cryotherapy or radical prostatec-
tomy? If clinicians do not consider these
factors, the agencies paying the bills likely will.
B In this regard, cryotherapy likely will be con-
sidered a highly cost-effective treatment.
There is the potential for cryotherapy (or
some other direct image-guided ablative tech-
nology) to become the predominant method of
prostate cancer treatment, based on its advan-
tages. The major criticism of the procedure in
the near future will be the relatively short
follow-up of data compared to more estab-
lished treatments. Numerous reports validate
the ability of cryotherapy to re-treat patients,
with no added morbidity, and superior results
in treating extracapsular disease.

c References
FIGURE 32.4. (A) Sagittal Tl-weighted magnetic res- 1. Villers A, McNeal IE, Freiha FS, et al. Multiple
onance imaging (MRI) showing a cryoprobe placed cancers in the prostate. Morphologic features
transperineally into the prostate of a dog. (B) Axial of clinically recognized vs. incidental tumors.
Tl-weighted image of the same dog's prostate gland Cancer 1992;70(9):2312-2318.
shows the cryoprobe in place in the anterior portion 2. George N. Therapeutic dilemmas in prostate
of the prostate. (C) Axial T1-weighted image shows cancer: justification for watchful waiting. Eur
freezing front (f) extending from the probe into the UroI1998;34(suppI3):33-36.
prostate. Note the asymmetric growth of the iceball, 3. Gondor MI, Soanes WA, Shulman S. Cryosurgi-
with minimal freezing occurring anteriorly due to the cal treatment of the prostate. Invest Urol1966;3:
anterior venous complex. 372-375.
410 G.Onik

4. Bonney WW, Fallon B, Gerber WL, et a1. Cryo- 18. Connolly JA, Shinohara K, Carroll P. Cryo-
surgery in prostatic cancer survival. Urology surgery for locally advanced prostate cancer.
1982;14:37-42. Semin Urol OncoI1997;15(4):244-249.
5. Onik GM, Cohen K, Reyes GD, et a1. Trans- 19. Miller RJ, Williams MJ, Sinner M, et a1. Percuta-
rectal ultrasound-guided percutaneous radical neous transperineal cryosurgical ablation of the
cryosurgical ablation of the prostate. Cancer prostate for the primary treatment of clinical
1993;72:1291-1299. stage C adenocarcinoma of the prostate. Urology
6. Pisters LL, von Eschenbach AC, Scott SM, 1995;44:676--679.
et a1. The efficacy and complications of salvage 20. Onik G, Narayan P, Brunelle R, et a1. Saline
cryotherapy of the prostate. J Urol 1997;921- injection into Denonvilliers' fascia during pros-
925. tate cryosurgery. Jnl Min Invas Therapy 2000;
7. Wong WS, Chinn DO, Chin M, et a1. Cryosurgery 9(6):423-427.
as a treatment for prostate carcinoma: results 21. Gould RS. Total cryosurgery of the prostate
and complications. Cancer 1997;79:963-974. versus standard cryosurgery versus radical
8. Cespedes RD, Pisters LL, von Eschenbach AC, prostatectomy: comparison of early results and
et a1. Long-term follow-up of incontinence and the role of transurethral resection in cryosurgery.
obstruction after salvage cryosurgical ablation of J UroI1999;162:1653-1657.
the prostate: results in 143 patients. J Uro11997; 22. Bahn DK, Lee F, Bandalament R, et a1. 7-year
157:237-240. outcomes in the primary treatment of prostate
9. Long JP, Bahn D, Lee F, et a1. 5 year retrospec- cancer. Urology 2002;60(2 suppl1):3-11.
tive multi-institutional pooled analysis of cancer- 23. Donnelly BJ, Salikan JC, Ernst DS, et a1.
related outcomes after cryosurgical ablation of Prospective trial of cryosurgical ablation of the
the prostate. Urology 2001;57(3):518-523. prostate: five year results. Urology 2002;60(4):
10. Onik GM, Cooper C, Goldberg HI, Moss AA, 645-649.
Rubinsky B, Christianson M. Ultrasonic char- 24. Salikan JC, Donnely B, Brasher P, et a1. Outcome
acteristics of frozen liver. Cryobiology 1984;21: and safety of transrectal US guided percuta-
321-328. neous cryotherapy for localized prostate cancer.
11. Onik G, Rubinsky B, Zemel R, et a1. Ultrasound J Vase Intervent Radiol 1999;10(2 pt 1):199-
guided hepatic cryosurgery in the treatment of 208.
metastatic colon carcinoma: preliminary results. 25. De La Taille A, Benson MC, Bagiella E, et a1.
Cancer 1991;67:901-907. Cryoablation for clinically localized prostate
12. Ravikumar TS, Kane R, Cady B, et a1. A five year cancer using an argon based system: complica-
study of cryosurgery in the therapy of liver tion rates and biochemical recurrence. BJU Int
tumors. Arch Surg 1991;126:1520-1524. 2000;85:281-286.
13. Steele G. Cryoablation in hepatic surgry. Semin 26. Robinson JW, Saliken JC, Donnelly BJ, et a1.
Liver Dis 1991;14(2):120-122. Quality of life outcomes for men treated with
14. Vaidya A, Hawke C, Tiguert R, et a1. Intra- cryosurgery for localized prostate cancer. Cancer
operative T staging in radical retropubic 86(9):1793-1800.
prostatectomy: Is it reliable? Urology 2001; 27. Onik G, Narayan P, Vaughan D, et a1. Focal
57(5):949-954. nerve-sparing cryosurgery for the treatment of
15. Partin AW, Yoo JK, Carter HB, et a1. The use primary prostate cancer: a new approach to pre-
of prostate specific antigen, clinical stage and serving potency. Urology 2002;60(1):109-114.
Gleason score to predict pathological stage in 28. Catalona WJ, Basler JW. Return of erections
men with localized prostate cancer. J Uro11993; and urinary continence following nerve sparing
150:110-115. radical retropubic prostatectomy. J Urol 1993;
16. Tawari A, Narayan P. Novel staging tool for 150(3):905-907.
localized prostate cancer: a pilot study using 29. Geary ES, Dendinger TE, Freiha FS, et a1. Nerve
genetic adaptive neural networks. J Urol 1998; sparing radical prostatectomy: a different view.
160(2):430-436. J UroI1995;154(1):145-149.
17. Eggleston JC, Walsh Pc. Radical prostatectomy 30. Sanchez-Ortiz RF, Broderick GA, Rovner ES,
with preservation of sexual function: pathologic et a1. Erectile function and quality of life after
findings in the first one hundred cases. J Urol interstitial radiation therapy for prostate cancer.
1985;134(6):1146--1148. Int J Impot Res 12(suppI3):S18-24.
32. Image-Guided Prostate Cryotherapy 411

31. Neerhut GJ, Wheeler T, Cantini M, et al. Salvage 34. Ghafar MA, Johnson CW, De La Taille, et al.
radical prostatectomy for radiorecurrent adeno- Salvage cryotherapy using an argon based
carcinoma of the prostate. J Urol 1988;140(3): system for locally recurrent prostate cancer after
544-549. radiation therapy: the Columbia experience.
32. Rainwater LM, Zincke H. Radical prostatec- J Urol2000;166(4):1333-1337.
tomy after radiation therapy for cancer of the 35. Tatsutani K, Rubinsky B, Onik GM, Dahiya R,
prostate: feasibility and prognosis. J Urol 1988; Narayan P. The effect of thermal variables on
140(6):1455-1459. frozen human primary prostatic adenocarci-
33. Bales GT, Williams MJ, Sinner M, et al. Short- noma cells. Urology 1996;48(3):441-447.
term outcomes after cryosurgical ablation of the 36. Lee F, Bahn DK, Badalament RA, et al. Cryo-
prostate in men with recurrent prostate cancer surgery for prostate cancer: improved glandular
following radiation therapy. Urology 1995;46: ablation by use of 6 to 8 cryoprobes. Urology
676-680. 1999;54(1):135-140.
33
Uterine Artery Embolization for
Fibroid Disease
Robert L. Worthington-Kirsch

Historical Background embolization (UAE) as a primary treatment for


fibroids (5,6).
Embolization of the uterine arteries has been Uterine artery embolization was introduced
the standard of care for management of acute in the United States by Bruce McLucas (a gyne-
bleeding after childbirth or after gynecologic cologist) and Scott Goodwin (an interventional
surgery since the late 1970s (1,2). Through the radiologist), both from UCLA Medical Center
1980s, apparently no one in either the inter- in 1996 (7). Francis Hutchins (gynecologist) and
ventional radiology or gynecologic communi- Robert Worthington-Kirsch (interventional
ties had thought of treating uterine fibroids by radiologist) introduced UAE to the eastern
embolization. This may have been due to the U.S. late in 1996 (8). Since then there has been
minimal interaction between interventional rapid spread of the procedure across the U.S.,
radiology and gynecology practices. Europe, and worldwide.
In the late 1980s, Jacques Ravina, a French
gynecologist, became interested in the possible
utility of embolization as a preemptive measure
before gynecologic surgery, such as myomec- Indications for Uterine
tomy. He was familiar with the utility of embo- Artery Embolization
lization for postoperative bleeding and decided
to investigate preoperative embolization; he The first indication for UAE is the presence of
hoped this would decrease intraoperative symptomatic fibroids. Fibroids (myomas of the
bleeding, as well as decrease the risk of post- uterus) are benign tumors of the uterus. They
operative hemorrhage. Preoperative emboliza- are extremely common, occurring in as many as
tion of the uterine arteries did indeed prove to 70% or more of women of childbearing age in
be useful to decrease perioperative bleeding some racial groups, such as African Americans
complications (3). (9,10).
In some cases, there was a delay between the Fibroids cause symptoms in only a portion of
embolization and the planned surgery of at women who have them, probably about 50%
least a few days, and in some cases a few weeks. (11). In general, fibroids that are not causing
Many of these patients experienced relief of any symptoms do not need to be treated. When
their fibroid-related symptoms from the embo- they do cause symptoms, these fall into one of
lization alone and refused to proceed with the three symptom groups.
planned gynecologic surgery. Ravina and his The first and most common symptom is
group first published their experience with a abnormal vaginal bleeding. Fibroids typically
small series of patients in 1995 (4), and went on cause both an increase in menstrual flow and
to continue their studies of uterine artery prolongation of the menses. This abnormal

412
33. Uterine Artery Embolization for Fibroid Disease 413

bleeding frequently is accompanied by worsen- number, and distribution of fibroids are not
ing of dysmenorrhea. The increase in bleeding particularly high, ranging from 8% to 46%
can be relatively mild or severe. In some (23,24). While we clearly need more informa-
patients the menorrhagia can be severe enough tion, the data currently available suggest that
that they are unable to leave their homes for fertility after UAE may be similar to fertility
several days of each menstrual cycle, or it can after myomectomy. This may be especially true
cause life-threatening anemia. Fibroids do not in those women who have multiple fibroids, and
have to be in a submucosal position to cause are generally regarded as poor candidates for
menorrhagia. myomectomy.
The second symptom complex is that of bulk- In the author's opinion, the desire to pre-
related symptoms. As the fibroids grow, they serve future fertility is not necessarily a con-
enlarge the uterus and cause pressure on traindication to UAE in patients who have
adjacent organs. The most common of these fibroid symptoms that would otherwise warrant
symptoms is from pressure on the bladder. treatment. Given the currently available
While urinary frequency, urgency, and nocturia data, UAE should not be considered for
are most common, occasionally a fibroid in women whose primary fibroid-related issue is
the lower uterine segment can cause pressure subfertility, unless an experienced fertility
on the bladder base and lead to episodes of surgeon feels that the patient is not a candidate
bladder outlet obstruction. Less commonly, the for myomectomy.
fibroid uterus can put pressure on the sigmoid
and rectum, causing a sensation of rectal pres-
sure and/or constipation, or on the sacrum Patient Selection
causing low back pain. The fibroid uterus can
also cause dyspareunia. Patient selection for UAE is a cooperative
The third symptom complex concerns prob- process between the interventional radiologist
lems with fertility. The relationship of fibroids and the referring gynecologist. The first issue is
and subfertility is complex and only partially to ensure that fibroids are indeed present and
understood (12,13). Most women with fibroid to exclude other significant pelvic pathology.
disease have normal fertility. It is clear that a This is done by a combination of the gyneco-
submucosal fibroid may distort the endometrial logic examination and imaging studies. The
cavity and interfere with implantation or pro- gynecologist should exclude other processes
gression of a pregnancy. However, fibroids else- such as endometriosis, pelvic inflammatory
where in the uterus also may have an untoward disease, and endometrial carcinoma. The author
impact on pregnancy. specifically requests endometrial evaluation [by
It is clear that UAE is an appropriate therapy hysteroscopy, endometrial biopsy, or dilatation
for women who have abnormal bleeding and/or and curettage (D&C)] in all women with exces-
bulk/pressure symptoms from their fibroid sive bleeding over age 40, and in all women with
disease (8,14-17). In women who want to main- abnormal bleeding patterns regardless of age.
tain future fertility, the utility of UAE is not so Imaging studies confirm the presence of
clear. The literature suggests that UAE for fibroids and exclude other pathology such as
other indications does not interfere with future undiagnosed adnexal masses or adenomyosis.
fertility (18). There are women who have Adenomyosis commonly can mimic the pre-
become pregnant after UAE (19-21), and most sentation of fibroid disease, and has been
have had normal pregnancy outcomes. There reported as a cause of clinical failure of UAE
are not yet enough data available to predict a (25). There has been work to investigate the
fertility rate after UAE with any confidence. possibility of treating adenomyosis by UAE
At the same time, the data regarding fertility (26-28), but the results have not been particu-
after myomectomy are not of particularly high larly encouraging.
quality (22). Fertility rates reported for women There are few absolute exclusion criteria for
having myomectomy without selection for size, UAE. These include pregnancy, evidence of
414 R.L. Worthington-Kirsch

active pelvic infection, and the presence of an through the vagina (30-32). The risk appears to
undiagnosed pelvic mass. Needless to say, a be 5% or less in the overall UAE population
history of life-threatening contrast allergy or (15,17). However, it may be as high as 25% in
other major contraindications to arteriography women who have a dominant submucosal
also may be exclusion criteria. When fibroids myoma (33). Sloughing of a fibroid usually is
coexist with adenomyosis, UAE may be suc- easily diagnosed and managed by the inter-
cessful, so long as the predominant process is ventional radiology/gynecology team. The
fibroid disease, but it may not be the best choice presence of a dominant submucosal myoma,
in patients with both fibroids and endometrio- therefore, is not a contraindication to UAE,
sis, since UAE will not relieve symptoms caused although the management team should be
by endometriosis. aware of the situation and potential effects.
Uterine artery embolization provides symp-
tomatic relief of either abnormal bleeding or
bulk/pressure symptoms in the majority of Technique
patients, regardless of initial uterine size (16).
In the author's opinion, there is no threshold or The technique for UAE is well described in the
limit for UAE in terms of uterine or fibroid size. literature (4,8,14,15,17). The uterine artery is
However, patients with large uteri do need to typically the first branch of the anterior division
be aware that their uterus may still be palpably of the internal iliac artery, although some
enlarged, even after significant volume reduc- anatomic variation has been described (34).
tion. Patients with large uteri who desire a "flat When catheterizing the uterine arteries, one
belly" are unlikely to be satisfied with the should be careful to avoid the cystic artery,
volume reduction afforded by UAE. which often shares a common trunk with the
Although not formally reported in the liter- uterine artery.
ature, there are two known cases in which an The catheter should be positioned in the
exophytic pedunculated fibroid was liberated main segment of the uterine artery. If possible,
into the peritoneal cavity after UAE, presum- the catheter tip should be distal to the origin of
ably because of necrosis of the stalk. In these the cervicovaginal branch of the uterine artery.
cases, the patients became symptomatic from However, this is not practical in most cases, as
the infarcted intraperitoneal myoma, and the cervicovaginal branch origin is often quite
laparotomy was required in both cases. A distal.
similar occurrence after surgery for fibroids Once the catheter tip is in position, the
has been reported (29). This should only be a uterine artery is embolized (Fig. 33.1). There
concern if the peduncle is quite narrow, and the are several options available to use for embolic
vast majority of exophytic fibroids, even those material. When UAE was first introduced as a
with a distinct peduncle, are not at risk for this therapy for fibroids, the protocol was simple.
occurrence. The author gives patients who are The uterine artery was selectively embolized
found to have fibroids on such narrow stalks with polyvinyl alcohol (PVA) particles, fol-
choices of: (a) deferring UAE for surgical lowed by "capping" with a plug of gelatin
therapy such as hysterectomy or myomectomy; sponge. The end point for embolization was to
(b) planning limited myomectomy to remove have a static column of contrast in the uterine
the pedunculated fibroid combined with UAE artery, with only a stump filling when the inter-
to manage the remainder of the patient's nal iliac artery was injected. The gelatin sponge
fibroid disease; or (c) doing a UAE with the cap was thought to both complete the occlusion
knowledge that there is a risk of the fibroid of the uterine artery and prevent PYA particles
being liberated into the abdomen, which can be from being drawn out of the uterine artery by
managed surgically if the need arises (although the Venturi effect, with resulting nontarget
the likelihood of that risk is not known). embolization.
There is a risk that fibroids may slough into Razavi and colleagues (35) have shown that
the endometrial cavity and need to pass UAE with PVA alone is effective without
33. Uterine Artery Embolization for Fibroid Disease 415

A B

c D

FIGURE 33.1. (A) Mapping pelvic arteriogram in a woman


with symptomatic fibroids in a 16-week-sized uterus. Note
the enlarged and tortuous uterine arteries (white aster-
isks). (B) Selective injection of the left uterine artery
before embolization. Note how the vessels surround the
dominant fibroid forming a perifibroid plexus. (C) After
embolization of the left uterine artery with PYA, there is
no further flow beyond the main uterine artery segment.
(D) Selective injection of the right uterine artery before
embolization. Note the collateral vessel filling across the
midline into the distribution of the just embolized left
uterine artery (black arrowhead). (E) After embolization
of the right uterine artery with PYA, there is no further
flow beyond the main uterine artery segment. (Images
courtesy of Tetsuya Katsumori, MD, Saiseikai Shiga Hos-
pital, Shiga Prefecture, Japan.) E
416 R.L. Worthington-Kirsch

evidence of nontarget embolization. The fibroid, while sparing the vessels that supply
author's experience has been that uterine arter- normal uterine tissue as much as possible. Given
ies embolized with PYA and gelatin sponge are these observations, what should the arterio-
usually completely obliterated at repeat arteri- graphic end point be? One should look for evi-
ography. This is seemingly in contradiction with dence that the flow dynamics of the uterus have
the standard teaching that gelatin sponge is a significantly changed, and that the blood flow to
"temporary agent". There must be a tissue reac- the perifibroid plexus has been interrupted. The
tion that stimulates the body to remove the angiographic signs of this are the following:
gelatin sponge, and thus it seems that this reac-
1. The appearance of new collaterals that
tion leads to durable occlusion of the uterine
were not present on initial injection of the
artery. Since UAE appears to destroy most (if
uterine artery. This may be the sudden appear-
not all) of the fibroids present at the time of the
ance of filling across the utero-ovarian anasto-
UAE, but does not change the underlying ten-
mosis, or the appearance of vessels cross-filling
dency of the uterus to form new fibroids, it is
to the opposite uterine artery.
important to preserve the main uterine artery
2. An increase in resistance of the uterine
segments after UAE, especially in younger
body vessels to further injection of contrast.
women, if only to preserve access for a second
This can be manifested by the beginning of
UAE at some point in the future. Given these
reflux in the uterine artery proximal to the
observations, this author has abandoned the use
catheter tip, dilation of the uterine artery when
of gelatin sponge for UAE in all patients.
pressure is exerted on the injection syringe, or
With the introduction of calibrated micro-
cessation of flow in the ascending ramus of the
sphere embolic agents, embolization has
uterine artery with contrast staining of the
become technically easier. The chief advantage
lower uterine segment.
of calibrated microspheres is that they are more
3. Occlusion of the main uterine artery.
uniformly sized than standard PYA prepara-
tions. This results in a more predictable level It appears that one can achieve the more
of embolization and minimizes the clogging of subtle of these end points more easily with cal-
both standard and microcatheters, which is a ibrated microspheres than with standard PYA
continuous problem with standard PYA prepa- preparations. However, it also should be noted
rations. This difference is so significant that a that there has been little or no observed differ-
simple dose equivalence between standard ence in the safety or clinical efficacy of UAE
PYA and microspheres cannot be determined using standard PVA compared to calibrated
(36). The two different preparations behave in microspheres.
markedly different ways. It has quickly become One of the most important technical con-
apparent that UAE with calibrated micro- siderations when performing UAE is the
spheres does not have to be performed to the possibility of flow across the utero-ovarian
end point of complete occlusion of the uterine anastomosis. If embolic material is allowed to
artery. This end point is difficult, if not impossi- reflux across this anastomosis into the ovary,
ble, to achieve with calibrated microspheres. there is risk of damage to the ovary (37,38). The
Comparison of embolization with PYA and severity of the risk to ovarian function is
microspheres in an animal model shows that unclear (39,40). Usually one can see flow across
PYA forms aggregates that often occlude the anastomosis, either on the initial injection
vessels much more proximally than would be of the uterine artery or developing as the
expected from the particle size used. This is embolization proceeds. In these cases, careful
apparently what leads to cessation of flow in the monitoring of the injection pressure during
main uterine arteries during UAE with stan- embolization can minimize or eliminate reflux
dard PVA preparations. of embolic material into the ovarian artery. It is
It must be remembered that the aim of em- important to recognize that the ovary itself is
bolization is not to occlude the main uterine several centimeters cephalad from the utero-
artery, but to occlude the vessels that supply the ovarian anastomosis, so reflux of material a
33. Uterine Artery Embolization for Fibroid Disease 417

very short way across this anastomosis should down into the uterus. Pelage et al (45) have
be harmless. On the other hand, there are times presented evidence suggesting that the branch
when there is significant flow into the uterus vessels into the ovary proper are typically 500
from the ovarian artery. This may occur as col- to 600/lm in diameter, so embolization with
lateral flow that is recruited by the demands of particles larger than 650 to 700/lm should
the fibroids (41,42), or in cases of anatomic vari- largely preclude any particles getting into the
ation in the uterine artery in which part of all ovary itself.
of the uterine artery branches are "replaced" by
Almost all patients develop significant
the ovarian artery (43). When there is signifi-
cramping after the procedure is completed,
cant flow to the uterus from the ovarian artery
similar to that seen with spontaneous degen-
that persists after embolization of the uterine
eration of a myoma. This usually requires IV
arteries, there is a good chance that the clinical
narcotics for pain control, as well as loading
results of DAE will be less than optimal. This is
doses of nonsteroidal antiinflammatory drugs
especially true when the ovarian artery supplies
(NSAIDs) such as ibuprofen. The acute post-
a portion of the uterus independent of the
procedure pain lasts 4 to 8 hours, after which
uterine artery. In these cases, supplemental
patients begin to feel better (46). By the
embolization of the ovarian artery should be
morning after DAE, all patients are discharged
considered, either at the same sitting (if the
from the hospital on oral medications.
informed consent for the DAE has included the
possibility of ovarian artery embolization) or at
a later date. Outcomes
The ovarian arteries usually both arise
directly from the aorta, although either (more All the published series of significant size
commonly the right) may originate from the report similar outcomes with DAE (6,15,17).
renal artery. Once catheterized, there are Technical success is reported at greater than
several choices of technique to embolize the 95%. Recovery is typically rapid, with the
ovarian artery: majority of patients returning to full activities
within 21 days of the procedure. Length of
1. Embolize the ovarian artery from its mid- hospitalization, return to work, and return to
portion, just as one would embolize the uterine normal activity levels all appear to be shorter
artery. This carries the highest risk of damaging with DAE than for patients who undergo sur-
the ovary. gical treatment for fibroids (47). The paradigm
2. Embolize the proximal ovarian artery for postprocedure management is the same as
with a larger particle such as a gelatin sponge for management of acute spontaneous fibroid
"torpedo" or a coil; this preserves the ovary, but infarction, that is, NSAIDs (such as ibuprofen),
would not get embolic material into the perifi- supplemented by narcotics (such as oxycodone)
broid vessels-rather relying on decreasing the as needed (48).
pressure head into the uterus as a supplement There are two management issues that have
to the embolization of the uterine arteries to to be watched for during the recovery phase.
infarct the fibroids. About 10% of patients develop a fever and
3. Advance a microcatheter down the malaise, sometimes accompanied by nausea
ovarian artery and into the anastomotic branch, and vomiting (15). This is similar to the post-
so that the embolization is into the uterus embolization syndrome seen after emboliza-
beyond the origin of the branches into the tion of other solid organs (49) or the post
ovary. This option requires not only superb ablation syndrome. In most cases, this resolves
microcatheter skills, but also a large amount of in 3 to 5 days, but can persist for a week or
procedure and fluoroscopy time (44). longer. Postembolization syndrome can be dif-
4. Embolize the ovarian artery with larger ferentiated from infection by the absence of
particles, which should then bypass the smaller worsening pelvic pain or purulent vaginal dis-
branches into the ovary itself and be washed charge.
418 R.L. Worthington-Kirsch

As has been mentioned above, a small patient, an infection has led to overwhelming
number of patients slough fibroids vaginally sepsis and death (53). There is one report of
after UAE (30-32). This may occur as early as patient mortality from pulmonary embolus the
several days after UAE, or as long as several day after UAE (54). Overall, the mortality risk
months after the procedure. Fibroid slough pre- associated with UAE is nearly an order of mag-
sents as the relatively abrupt onset of crampy nitude lower than the mortality risk associated
abdominal pain and odorous vaginal discharge. with hysterectomy for fibroids (54). The overall
These patients should be treated with oral risk of complication seems to be lower for UAE
antibiotics to prevent ascending infection. In than for hysterectomy (47,55).
most cases, the sloughed fibroid will pass spon-
taneously within 24 to 36 hours, with resolution
of symptoms. Although the fibroid may pass Future Directions
spontaneously beyond this period, a longer
delay increases the risk for ascending infection. Much about UAE is currently unknown. Proj-
The author routinely requests that the referring ects are ongoing to formally compare UAE to
gynecologist perform a dilation and evacuation surgical therapies such as hysterectomy and
(D&E) to remove the sloughed fibroid if it has myomectomy, and to gather long-term outcome
not passed within 48 hours of the onset of data. New embolization materials are being
symptoms. developed, and refinements to both the tech-
The majority of women experience no inter- nique of embolization and patient management
ruption in their menstrual cycle after UAE. are evolving rapidly. Many questions about the
There is risk for amenorrhea after UAE, par- place of UAE for younger women, particularly
ticularly for women over 45 years of age those who want to preserve fertility, remain and
(50-52). Approximately 90% of women have are a ripe field for further research.
improvement or normalization of menstrual
flow after UAE (6,8,15,17).
Uterine and fibroid volumes decrease after Conclusion
UAE, although the degree of volume reduction
is variable. At 3 months after UAE, 35% to Uterine artery embolization has been estab-
50% reduction in overall uterine volume is lished as a valuable ablative therapy for the
typical (6,15,17). Volume reduction continues in management of fibroid disease. It affords dura-
many women for at least 12 months after UAE; ble therapy for fibroid symptoms by a pro-
80% to 85% of women who have pressure cedure that is less invasive than traditional
symptoms caused by fibroid bulk report surgical therapies. The advantages of UAE over
improvement in those symptoms within 3 surgical treatment include shorter hospitali-
months of UAE (6,15,17). This is independent zation, lower complication risk, and faster
of the presence of, or change in, abnormal recovery.
bleeding (16). Symptomatic relief (both from
menorrhagia and bulk-related symptoms) after
UAE appears to be lasting. References
1. Heaston OK, Mineau DE, Brown BJ, Miller FJ.
Complications Transcatheter arterial embolization for the con-
trol of persistent massive puerperal hemorrhage
after bilateral surgical hypogastric artery
Complications of UAE have proven to be rare. ligation. AJR 1979;133:152-154.
Infection can develop, particularly in the setting 2. Oliver JA, Lance JS. Selective embolization to
of a sloughing fibroid that is not managed control massive hemorrhage following pelvic
aggressively; this may lead to urgent or emer- surgery. Am J Obstet GynecoI1979;135:431-432.
gent hysterectomy. The incidence of this occur- 3. Ravina JH, Merland JJ, Herbreteau 0, Houdart
rence is less than 1 in 200 cases (17). In one E, Bouret JM, Madelenat P. Embolisation pre-
33. Uterine Artery Embolization for Fibroid Disease 419

operatoires des fibromes uterins. Presse Med on menses and pregnancy. Radiology 1996;201:
1994;23:1540. 179.
4. Ravina JH, Herbreteau D, Ciraru-Vigneron N, 19. Ravina JH, Ciraru-Vigneron N, Aymard A,
et al. Arterial embolisation to treat uterine LeDref 0, Merland J1. Pregnancy after embo-
myomata. Lancet 1995;346:671-672. lization of uterine myoma: report of 12 cases.
5. Ravina JH, Aymard A, Ciraru-Vigneron N, et al. Fertil Steril 2000;73:1241-1243,
Embolisation arterielle particulaire: un nouveau 20. McLucas B, Goodwin S, Adler L, Rappaport A,
traitement des hemorragies des leiomyomes Reed R, Perella R. Pregnancy following uterine
uterins. Presse Med 1998;27:299-303. artery embolization. Int J Gynecol Obstet 2001;
6. Ravina JH, Ciraru-Vigneron N, Aymard A, 74:1-7.
Ferrand J, Merland JJ. Uterine artery embolisa- 21. Pelage Jp' Walker W1. Uterine artery embolisa-
tion for fibroid disease: results of a 6 year study. tion for symptomatic fibroids and pregnancy.
Min Invas Ther Allied TechnoI1999;8:441--447. J Vase Intervent Radiol 2002;13:S65.
7. McLucas B, Goodwin Sc. A fibroid treatment 22. Myers ER, Barber MD, Couchman GM, et al.
with promise-and a catch. Obstet Gynecol Evidence report: management of uterine
Manag 1996;8:53-57. fibroids. US Agency for Healthcare Research
8. Worthington-Kirsch RL, Hutchins FL, Popky and Quality (AHRQ), Contract No. 290-97-0024,
GL. Uterine artery embolization for the man- Task Order 4,2000.
agement of leiomyomas: quality of life assess- 23. Bernard G, Darai E, Poncelet C, Benifla JL,
ment and clinical response. Radiology 1998;208: Madelenat P. Fertility after hysteroscopic myo-
625--629. mectomy: effect of intramural myomas associ-
9. Cramer SF, Patel D. The frequency of uterine ated. Eur J Obstet Gynecol Reprod Bioi 2000;
leiomyomas. Am J Clin PathoI1990;94:435-438. 88:85-90.
10. Schwartz SM. Epidemiology of uterine leiomy- 24. Sudik R, Harsch K, Steller J, Daume E. Fertility
omata. Clin Obstet Gynecol 2001;44:316-326. and pregnancy outcome after myomectomy in
11. Greenberg MD, Kazamel TIG. Medical and sterility patients, Eur J Obstet Gynecol Reprod
socioeconomic impact of uterine fibroids. Obstet Bioi 1996;65:209-214.
Gynecol Clin North Am 1995;22:625--636. 25. Smith SJ, Sewall LE, Handelsman A, A clinical
12. Forman RG, Reidy J, Nott V, Braude P. Fibroids failure of uterine artery embolization due to
and fertility. Min Invas Ther Allied Technol1999; adenomyosis. J Vase Intervent Radiol 1999;10:
8:415--419. 1171-1174.
13. Rice JP, Kay HH, Mahony BS. The clinical sig- 26. Siskin GP, Tublin ME, Stainken BF, Dowling K,
nificance of uterine leiomyomas in pregnancy. Dolen EG. Uterine artery embolization for the
Am J Obstet GynecoI1989;160:1212-1215. treatment of adenomyosis. AJR 2001;177:297-
14. Goodwin SC, Vedantham S, McLucas B, Forno 302.
AE, Perella R. Uterine artery embolization for 27. Kim MS, Wan JW, Lee DY, Ahn CS, Uterine
uterine fibroids: results of a pilot study. J Vase artery embolization for adenomyosis without
Intervent RadioI1997;8:517-526. fibroids. Clin Radiol 2004;59:520-526.
15. Hutchins FL,Worthington-Kirsch RL, Berkowitz 28. Katsumori TK, Ueda M, Nishino T, Bamba M,
RP. Selective uterine artery embolization as Katsumori T, Kushima R. Cellular changes fol-
primary treatment for symptomatic leiomy- lowing uterine artery embolization for the treat-
omata uteri: a review of 305 consecutive cases. ment of adenomyosis. Cytopathology 2001;12:
J Am Assoc Gynecol Laparosc 1999;6:279- 270-272.
284. 29. Worthington-Kirsch RL, Hutchins FL. Retained
16, Delaney ML, Worthington-Kirsch RL, Hutchins myoma fragment after LASH procedure-case
FL, Berkowitz RP. Uterine artery embolisation report and imaging findings (letter). Clin Radiol
for the management of myomata in patients 2001;56:777-778.
without complaints of menorrhagia, Min Invas 30. Berkowitz RP, Hutchins FL, Worthington-Kirsch
Ther Allied TechnoI1999;8:455--458. RL. Vaginal expulsion of submucosal fibroids
17. Spies JB, Ascher SA, Roth AR, Kim J, Levy EB, following uterine artery embolization: a report
Gomez-Jorge 1. Uterine artery embolization for of three cases, J Reprod Med 1999;44:373-
leiomyomata. Obstet GynecoI2001;98:29-34. 376.
18. Stancato-Pasik A, Mitty HA, Richard HM III, 31. Abhara S, Spies JB, Scialli AR, Jha RC, Lage JM,
Eshkkar NS. Obstetric embolotherapy: effect Nikolic B. Transcervical expulsion of a fibroid
420 R.L. Worthington-Kirsch

as a result of uterine artery embolization for 42. Matson M, Nicholson A, Belli AM. Anastamoses
leiomyomata. J Vasc Intervent Radiol 1999;10: of the ovarian and uterine arteries: a potential
409-411. pitfall and cause of failure of uterine emboliza-
32. Jones K, Walker WJ, Sutton C. Sequestration and tion. Cardiovasc Intervent Radiol 2000;23:393-
extrusion of intramural fibroids following arter- 396.
ial embolization: a case series. Gynaecol Endosc 43. Worthington-Kirsch RL, Walker WJ, Adler L,
2000;9:300-313. Hutchins FL. Anatomic variation in the uterine
33. McLucas B. Management of the post- arteries: a cause of failure of uterine artery
embolization patient. Presented at New Trends embolisation for the management of sympto-
in Myoma Management Including Uterine matic fibroids. Min Invas Ther Allied Technol
Fibroid Embolization, St. Thomas' Hospital, 1999;8:397-402.
London, England, 1 June 2001. 44. Andrews RT, Bromley PJ, Pfister ME. Successful
34. Gomez-Jorge J, Kyoung A, Spies JB. Uterine embolization of collaterals from the ovarian
artery anatomy relevant to uterine leiomyomata artery during uterine artery embolization for
embolization. Presented at SCVIR Uterine fibroids: a case report. J Vasc Intervent Radiol
Artery Embolization (UAE) Conference, Wash- 2000;11:607-610.
ington, DC, 14 October 2000. 45. Pelage JP, Laurent A, Wassef M, et a1. Uterine
35. Razavi MK, Rhee J, Sze DY, Kee ST, Semba CP, artery embolization in sheep: comparison of
Dake MD. Recanalization of uterine arteries acute effects with polyvinyl alcohol and cali-
after embolization for symptomatic leiomyomas: brated microspheres. Radiology 2002;224:436-
evidence on MRA. J Vasc Intervent Radio12000; 445.
11:S286. 46. Worthington-Kirsch RL, Koller NE. Time course
36. Levine A, Kim D, Termin P, Worthington-Kirsch of pain after uterine artery embolization for
RL. Comparison of embosphere and PVA fibroid disease. Medscape Women's Health
embolization agents in a porcine renal model. Journal 2002;7:http://www.medscape.com/
Presented at Cardiovascular and Interventional viewarticle/430765.
Radiological Society of Europe (CIRSE) 2001 47. Worthington-Kirsch RL. Uterine artery em-
Annual Meeting and Postgraduate Course, bolization for leiomyomata using embosphere
Gothenburg, Sweden, 22-26 September 2001, microspheres: interim results of a phase II
(manuscript in preparation). comparative study. Presented at New Trends
37. Ryu RK, Chrisman HB, Omary RA, et a1. The in Embolotherapy Symposium sponsored by
vascular impact of uterine artery embolization: Biosphere Medical associated with CIRSE 2002,
prospective sonographic assessment of ovarian 5-9 October 2002, Luzerne, Switzerland.
arterial circulation. J Vasc Intervent Radiol 48. Rapkin Al Pelvic pain and dysmenorrhea In:
2001;12:1071-1074. Berek JS, ed. Novak's Gynecology. 12th ed. Bal-
38. Stringer NH, Grant T, Park J, Oldham L. Ovarian timore: Williams & Wilkins, 1996: 405.
failure after uterine artery embolization for 49. Hemingway AP. Complications of embolother-
treatment of myomas. J Am Assoc Gynecol apy. In: Kadir S, ed. Current Practice of Inter-
Laparosc 2000;7:395-400. ventional Radiology. Philadelphia: B.C. Decker,
39. Chrisman HB, Saker MB, Ryu RK, et a1. The 1991: 104-109.
impact of uterine fibroid embolization on 50. Chrisman HB, Saker MB, Ryu RK, et a1. The
resumption of menses and ovarian function. impact of uterine fibroid embolization on
J Vasc Intervent Radio12000;11:699-703. resumption of menses and ovarian function.
40. Spies JB, Roth AR, Gonsalves SM, Murphy- J Vasc Intervent Radio12000;11:699-703.
Skrzniarz KM. Ovarian function after uterine 51. Spies JB, Roth AR, Gonsalves SM, Murphy-
artery embolization for leiomyomata: assess- Skrzniarz KM. Ovarian function after uterine
ment with use of serum follicle stimulating artery embolization for leiomyomata: assess-
hormone assay. J Vasc Intervent Radiol 2001;12: ment with use of serum follicle stimulating
437-442. hormone assay. J Vasc Intervent Radiol 2001;12:
41. Nikolic B, Spies JB, Abhara S, Goodwin Sc. 437-442.
Ovarian artery supply of uterine fibroids as a 52. Tulandi T, Sammour A, Valenti D, Child TJ, Seti
cause of treatment failure after uterine artery L, Tan SL. Ovarian reserve after uterine artery
embolization: a case report. J Vasc Intervent embolization for leiomyomata. Fertil Steri12002;
Radiol1999;10:1167-1170. 78:197-198.
33. Uterine Artery Embolization for Fibroid Disease 421

53. Vashisht A, Studd JWW, Carey AH, et a1. Fibroid 55. Walker WJ, Worthington-Kirsch, RL. Fatal
embolisation: a technique not without significant septicemia after uterine fibroid embolisation
complications. Br J Obstet Gynaecol 2000;107: (letter). Lancet 1999;354(9191):1730.
1166-1170. 56. Spies JB, Cooper JM, Worthington-Kirsch RL,
54. Lanocita R, Frigerio LF, Patelli G, et a1. A fatal Lipman JC, Benenati JF, McLucas B. Uterine
complication of percutaneous transcatheter artery embolization (UAE) using embospheres:
embolization for the treatment of fibroids. Pre- initial results of a phase II comparative study.
sented at the 2nd International Symposium on Presented at SCVIR 2002, 27th Annual Scientific
Embolization of Uterine Myomata, the Society Meeting, 6-11 April 2002, Baltimore.
for Minimally Invasive Therapy, 11th Interna-
tional Conference, Boston, 16-18 September
1999.
34
Applications of Cryoablation
in the Breast
John C. Rewcastle

The use of cryoablation in the breast has been The majority of the iceball experiences lower
anecdotally investigated since the mid-1800s freezing rates that lead to cellular dehydration
(1). Recently, however, more concerted and that result from the osmotic imbalance [5, 6].
structured research has been conducted that This mechanism has been termed "solution
builds upon the modern understanding of cryo- effects" and there exist several hypotheses, but
biology and several advances in technology. no consensus, as to its physical basis. Result-
Freezing and thawing of tissue result in ing solute concentration alone has been held
necrosis. It is important to note that cryother- responsible for the damage, as has attainment
apy injures not only individual cells at the time of a critical minimum volume shrinking, beyond
of therapy (direct damage), but also the tissue which the membrane is damaged (6).
as a whole by impairing the microvasculature Direct injury does not completely destroy all
(indirect damage) (2). Unlike radiation, the cells. In the hours and days following cryoabla-
ability of cryotherapy to ablate tissue is depen- tion, indirect damage occurs. Microvasculature
dent not on the nuclear characteristics of indi- endothelial cells also are damaged by the direct
vidual cells, but rather on the exposure of the injury mechanisms, resulting in ischemia that
entire tumor volume to a lethal freezing greatly enhances cell kill within the iceball. This
process. Cellular survival during cryoablation may be the dominant killing mechanism during
depends not only on the freezing and thawing cryosurgery (6). In vitro studies have shown
rates, but, most importantly, on the lowest tem- that induced apoptosis also may playa role (7).
perature reached and the hold time at subzero Enhancing apoptosis with adjuvant chemother-
temperatures (3). During cryoablation, ice, apy may indeed lead to a significant increase
which is essentially pure water, initially forms in in vivo cryoinjury, and investigations are
outside the cell, increasing the extracellular ongoing (8).
concentration. It does not form initially inside Several reports have noted that the integrity
the cells as the lipid membrane blocks ice of some structures is not compromised by expo-
crystal growth, resulting in an osmotic imbal- sure to cryogenic temperatures. For example,
ance. Direct cellular injury can result and is due the renal collecting system is not damaged
to two damage mechanisms: intracellular ice during cryotherapy even when exposed to very
formation (IIF) and solution effects. low temperatures unless it is physically punc-
Freezing rates are high enough within only a tured with a probe, and transmural lesions are
few millimeters of the cryoprobe to induce IIF created routinely during cardiac cryoablation
(4,5). Although IIF is usually a lethal event without impairing the integrity of the heart
associated with irreversible membrane damage, wall (9,10). There also exists some evidence of
it is not known if cell death is a cause or a result an immunologic response to cryoablation, but
of IIF, and several mechanistic theories exist (6). experimental results have been inconsistent

422
34. Applications of Cryoablation in the Breast 423

and its existence, let alone significance, remains lumpectomy, and the use of cryoablation as a
controversial (2). primary breast cancer therapy.
The other significant advance in cryoablation
has been the recent introduction of high-
pressure gas cryoablation systems that exploit Cryoablation for the Treatment
the Joule-Thompson (J-T) effect. The J-T
effect, a behavior characteristic of high- of Biopsy-Proven
pressure gas, predicts that gas changes temper- Fibroadenomas
ature as it expands through a narrow port into
a lower pressure zone. This is a constant Approximately 10% of women experience a
enthalpy expansion that, in the case of argon fibroadenoma in their lifetime (11). Fibroade-
gas, results in rapid GOoling to the boiling point nomas are considered aberrations of normal
of argon gas (-186°C). To accomplish this, high- breast tissue rather than benign tumors (12),
pressure (3000psi) ambient temperature argon and consist of combined proliferation of epithe-
gas is circulated to the cryoprobe tip where it lial and connective tissue elements, usually
expands rapidly as it drops to room pressure developing in the lobular region (11). Most
(15 psi). The expanded gases are circulated back fibroadenomas stop growing after becoming 2
to the cryogenic unit through the larger outer to 3 cm in size, with 15% spontaneously regress-
lumen of the cryoprobe and the supply hose. ing and 5% to 10% progressing (11,13).
The venting of used gas, usually into the room, Historically, surgical resection has been the
occurs at the cryogenic machine. Freezing is treatment of choice for breast fibroadenoma.
controlled by a computer-modulated gas regu- The advantages include providing a definitive
lator that opens and closes the gas flow supply diagnosis while eliminating the source of
to the cryoprobe. patient anxiety, and alleviating the need for
Under the J-T effect, some gases, such as follow-up monitoring, which relies on long-
helium, warm rather than cool when expanded. term patient compliance. Drawbacks to exci-
Accordingly, helium can be incorporated into sion include the cost and morbidity (cosmetic
the cryogenic system to act as a rapidly respon- or ductal damage), which may be unnecessary
sive cryoprobe heater as it expands to ambient in a benign lesion that may resolve sponta-
pressure. It is noteworthy that both argon and neously remit (11,14). There is also the aware-
helium are inert and therefore, safe gases. ness that resection of every fibroadenoma
The gas systems provide finely adjustable and would place a huge burden on the health care
rapid system responses (seconds) to user input. system (14).
This is in comparison to liquid nitrogen systems In the past decade there has been a growing
that had a lag time of up to 2 minutes. They also acceptance of conservative management (i.e.,
support rapid conversion from cooling to observation and monitoring) with a corre-
heating, effected through computer-automated sponding push to reserve resection for limited
switching of argon to helium. The gas cryo- circumstances. Unfortunately, definitive diag-
probes, which can modulate between -186°C nosis is seldom certain with conservative man-
and +40°C in about 30 seconds, do not demon- agement (15). Some physicians have expressed
strate any clinically perceptible promotion of great reluctance about letting a lump remain in
freezing during the interval switch. a breast in a patient who will most likely be lost
It is the understanding of cryobiology as well to follow-up, so that regular monitoring cannot
as the introduction of advanced argon gas- take place (16). Further, there remain abundant
based cryomachines operated under ultrasound indications in the literature for resection. Some
guidance that allow for the accurate removal of of these indications include an increase in
heat from a target tissue. This has led to inves- lesion size, lack of regression by age 35 (11), a
tigations of cryoablation for the treatment of higher risk of phylloides tumor in masses >3cm
biopsy-proven fibroadenomas, the use of a (17), multiple fibroadenomas (14), and a mass
cryoprobe as a localization tool to assist in that is psychologically disturbing to the patient
424 Ie. Rewcastle

FIGURE 34.2. Ultrasound visualization of a cryo-


probe placed in the center of the breast lesion.
FIGURE 34.1. Ultrasound visualization of a breast
lesion.
and exposes it to temperatures of -40°C or less.
A double freeze-thaw cycle is used routinely.
(18). Further, with epidemiologic evidence that Numerous studies have shown that tissues
the mean age of fibroadenoma diagnosis is the exposed to -40°C or less during two subsequent
mid-20s and the mean age of diagnosis of freeze-thaw cycles will, without question, be
fibroadenoma containing a carcinoma is 43 ablated (21). Figure 34.3 shows the hyperechoic
years, Deschenes et al (19) recommend that all rim of an iceball as observed on ultrasound.
fibroadenomas diagnosed in women older than Note that only the proximal edge of the iceball
30 years be excised. is visible as near 100% sonographic reflection
Cryoablation presents as a minimally inva- occurs at the interface between frozen and
sive treatment option for women with biopsy- unfrozen tissue. Figure 34.4 demonstrates the
proven fibroadenomas. In fact, cryoablation relation between the ultrasound probe and the
seems to be ideally suited for this application as iceball. In the situation shown one would
it has a proven ability to target and ablate tissue observe an ultrasound image akin to that in
in situ, does not require physical removal of Figure 34.3. If the lesion is near the skin surface,
the fibroadenoma or treatment of large saline can be injected between the lesion and
amounts of surrounding normal tissue, and skin to increase their physical separation. There
requires only a 3- to 4-mm incision for the are no instances in which the skin was damaged
insertion of a cryoprobe. The combination of by an approaching iceball. Discomfort is tran-
these factors has led to several clinical trials sient in all patients and rarely requires medica-
assessing the ability of cryoablation to effec-
tively treat fibroadenomas.
Kaufman et al (20) reported a series of 50
patients with 57 biopsy proven fibroadenomas.
Cryoablation was performed in the operating
room only for the first seven patients who
underwent intravenous sedation, after which all
patients were treated comfortably in the office
with local anesthesia. The size of the treated
lesions varied from 7 to 42 mm.
The procedure consists of locating the lesion
on ultrasound (Fig. 34.1), then advancing a cryo-
probe into the center of the lesion (Fig. 34.2).
Using a treatment algorithm based on lesion FIGURE 34.3. Ultrasound visualization of an iceball.
size, an iceball forms and engulfs e.ach lesion The hyperechoic rim is the proximal iceball edge.
34. Applications of Cryoablation in the Breast 425

almost always accomplish diagnosis, but are


very poor at ensuring a tumor-free margin in
the specimen. Although the definition of a
"clear margin" remains controversial, surgical
margin involvement with breast cancer almost
always results in obligatory reexcision or mas-
tectomy. For wire-localized procedures, the
reexcision rate (requiring return of the patient
to the operating room for a second procedure)
often is reported to be between 40% and 60%
(23-30).
The essence of a cryoprobe-assisted lumpec-
tomy is that the tumor and a margin are
enclosed by an iceball localized prior to surgi-
cal excision. Under ultrasound guidance a cryo-
FIGURE 34.4. Drawing of the breast surface, ultra- probe is inserted into the center of the tumor.
sound probe, and iceball. This situation would The cryoprobe is then engaged until an iceball
produce an ultrasound image like that in Figure 34.3. is created that encloses not only the entire
tions other than acetaminophen or ibuprofen. tumor, but also a margin. Once large enough,
As expected, localized swelling and skin ecchy- the operation of the cryoprobe can be modified
mosis are observed, but typically resolve within such that the iceball no longer grows, but main-
3 weeks. Breast appearance returns to baseline tains a static size. The iceball enclosing the
universally within 6 weeks. tumor is then surgically excised, which may
Following initial swelling progressive absorp- prove to be a superior localization methodol-
tion of the fibroadenoma is observed on ogy than needle wire localization.
ultrasound. At 6 months the average volume Tafra et al (31) report a pilot study of 24
decrease is 65% and 92% at 12 months. Patient patients who underwent a cryoprobe-assisted
satisfaction is excellent. lumpectomy for their ultrasound visible breast
In a similar study Littrup et al (22) treated 42 cancers. All lesions were localized successfully
biopsy-proven fibroadenomas in 27 patients. and the negative margin rate for patients
Minimal discomfort was reported with the use with a margin greater than 6mm was 5.6%.
of local anesthesia. No significant complications Although this study is neither large nor com-
occurred. Again, patient satisfaction was excel- parative the results are compelling compared to
lent. Fibroadenoma volume reduction at 12 the reported positive margin rate in the litera-
months was observed under ultrasound to be ture. As a follow-up, a multicenter randomized
73%. trial is planned that compares cryoprobe-
assisted localization to needle wire localization.
Cryoprobe-Assisted
Lumpectomy
Cryoablation as a Primary
Over the past 30 years various techniques have Therapy for Breast Cancer
been util.ized for the preoperative localization
of nonpalpable or barely palpable breast There is very little in the literature regarding
lesions, the most common of which is guidewire the use of cryoablation as a primary therapy
localization using mammographic or ultrasono- for breast cancer. A single case study of in situ
graphic guidance. The purpose of needle- cryoablation of breast cancer was published by
localized procedures was always twofold: (1) Staren et al (32). There are,however, two similar
diagnosis, and (2) excision of a malignancy with studies in which patients underwent cryoabla-
an adequate tumor-free margin of normal tion prior to surgical excision. Pfleiderer et al
tissue. Needle-wire localization procedures (33) used a 3-mm cryoprobe to ablate 16 lesions
426 Ie. Rewcastle

in 15 patients.There were no complications asso- positive margin rate following lumpectomy. If


ciated with the cryoablation procedure and all these results are substantiated in the planned
patients underwent resection within 5 days. The randomized trial, cryoprobe-assisted lumpec-
authors concluded that the invasive components tomy could replace wire localization as the
of small tumors can be treated effectively with most common localization method used during
cryoablation, but remnant ductal carcinoma in lumpectomy.
situ (DCIS) components not observed prior to Initial investigations of cryoablation as a
treatment may be problematic. Further, they primary therapy for breast cancer show that
recommend that multiple cryoprobes be used if there are potential pitfalls. The maximum lesion
the lesion is greater than 15 mm. size that can be effectively treated is limited by
In a similar study, Sabel et al (34) reported on the number and size of cryoprobes. Further,
29 patients with ultrasound-visible breast DCIS components undetected prior to therapy
cancer <2.0 cm who underwent a double freeze- may be of concern. These two issues are cer-
thaw cycle cryoablation utilizing a single tainly surmountable by careful treatment plan-
cryoprobe. The cryoprobe design changed ning and diagnosis.
mid-study from a diameter of 2.4 mm to 2.7 mm. Cryoablation has the ability to ablate in a
Subsequent standard surgical resection was uniform and confluent manner without remov-
performed on all patients 1 to 4 weeks post- ing a breast mass or leaving an unsightly scar.
cryoablation. The procedure was very well tol- This results in a therapy that is effective, safe,
erated with no complications and no need for and attractive to the patient due to minimal
narcotic pain medications. Pathologic examina- quality-of-life impact. The use of cryoablation
tion found that all tumors <1.0cm were 100% for the treatment of both benign and malignant
ablated; 100% ablation also was achieved in breast conditions is sure to increase in the
invasive ductal carcinoma without a significant future.
DCIS component «25% on biopsy) when the
carcinoma size was between 1.0 and 1.5 cm.
These results have formed the basis of a Acknowledgments. The author would like to
planned multicenter trial. It is expected that acknowledge the assistance of Trena Depel
additional independent trials regarding cryoab- and Mark Rose in the preparation of this
lation as a primary therapy for breast cancer manuscript.
also will begin shortly.
Both these studies clearly demonstrate the
ability of cryoablation to effectively treat a References
target volume. The size and number of probes 1. Arnott I Practical illustrations of the remedial
will dictate the maximum size of lesion that can efficacy of a very low or anesthetic temperature.
be treated. Lancet 1850;2:257-259.
2. Hoffmann NE, Bischof Je. The cryobiology of
cryosurgical injury. Urology 2002;60(2 suppll):
Conclusion 40-49.
3. Smith DJ, Fahssi WM, Swanlund DJ, Bischof Je.
A parametric study of freezing injury in AT-1 rat
Cryoablation is being used in several different
prostate tumor cells. Cryobiology 1999;39(1):13-
ways in the management of benign and malig-
28.
nant breast tumors. Results from the treatment 4. Bischof JC, Smith D, Pazhayannur PV, Manivel
of fibroadenomas are encouraging and cryo C, Hulbert J, Roberts KP. Cryosurgery of
now represents a safe and effective alternative dunning AT-1 rat prostate tumor: thermal,
with minimal cosmetic impact for women who biophysical, and viability response at the cellular
choose to have definitive action taken. The neg- and tissue level. Cryobiology 1997;34(1):42-69.
ative margin rates from the cryoprobe-assisted 5. Rewcastle JC, Muldrew K, Sandison GA. In vitro
lumpectomy pilot study are significant as they injury mapping for single cryoprobe cryosurgery.
represent almost a tenfold reduction in the Cryobiology 2001;43:322.
34. Applications of Cryoablation in the Breast 427

6. Gage AA, Baust 1. Mechanisms of tissue injury the human prostate during cryosurgery with
in cryosurgery. Cryobiology 1998;37(3):171-186. correlation to histopathologic outcomes.
7. Baust 1M, Van B, Baust lG. Cell viability Urology 2000;55:547-552.
improves following inhibition of cryopreserva- 22. Littrup Pl, Freeman-Gibb L, Andea A, et al.
tion-induced apoptosis. In Vitro Cell Dev BioI Cryotheapy for breast fibroadenomas. Radiol-
Anim 2000;36(4):262-270. ogy 2005;234:63-72.
8. Clarke DM, Baust 1M, Van Buskirk RG, Baust 23. Pittinger TP, Maronian NC, Poulter CA, Peacock
lG. Chemo-cryo combination therapy: an lL. Importance of margin status in outcome
adjunctive model for the treatment of prostate of breast-conserving surgery for carcinoma.
cancer. Cryobiology 2001;42(4):274-285. Surgery 1994;116:605-608.
9. Lustgarten DL, Keane D, Ruskin 1. Cryothermal 24. Blair SL, O'Shea KE, Orr RK. Surgeon variabil-
ablation: mechanism of tissue injury and current ity in treating nonpalpable breast cancer: surgi-
experience in the treatment of tachyarrhythmias. cal oncology as a value-added specialty. Ann
Prog Cardiovasc Dis 1999;41(6):481-498. Surg OncoI1998;5:28-32.
10. Sung GT, Gill IS, Meraney AM. Effect of inter- 25. Velanovich V, Lewis FR lr, Nathanson SD,
ventional cryoinjury to the renal collecting et al. Comparison of mammographically guided
system (abstract). 1 Urol 2000;163:113. breast biopsy techniques. Ann Surg 1999;229:
11. Sperber F, Blank A, Metser U, et al. Diagnosis 625-630.
and treatment of breast fibroadenomas by ultra- 26. Tartter PI, Kaplan 1, Bleiweiss I, et al. Lumpec-
sound-guided vacuum-assisted biopsy. Arch Surg tomy margins, reexcision, and local recurrence of
2003;138:796-800. breast cancer. Am 1 Surg 2000;179:81-85.
12. Houssami N, Cheung MNK, Dixon M. Fibroade- 27. Luu HH, Otis CN, Reed WP lr, Garb lL, Frank
noma of the breast. Med 1 Aust 2001;174:185- lL. The unsatisfactory margin in breast cancer
188. surgery. Am 1 Surg 1999;178:362-366.
13. Pick PW, Iossifide IA. Occurrence of breast car- 28. lardines L, Fowble B, Schultz D, et al. Factors
cinomas within a fibroadenoma: a review. Arch associated with a positive reexcision after exci-
Pathol Lab Med 1984;108:590-593. sional biopsy for invasive breast cancer. Surgery
14. Greenberg R, Skornick Y, Kaplan 0. Manage- 1995;118:803-809.
ment of breast fibroadenoma. 1 Gen Intern Med 29. Chinyama CN, Davies lD, Rayter Z, et al.
1998;13:640-645. Factors affecting surgical margin clearance in
15. Takei H, Lino Y, Horiguchi 1, et al. Natural screen detected breast cancer and the effect of
history of fibroadenomas based on the correla- cavity biopsies on residual disease. Eur 1 Surg
tion between size and patient age. lpn 1 Clin OncoI1997;23:123-127.
OncoI1999;29:8-10. 30. Rahusen FD, Taets van Amerongen AHM, Van
16. Alle K. Conservative management of fibroade- Diest Pl, Borgstein Pl, Bleichrodt RP, Meijer S.
noma of the breast: author's reply. Br 1 Surg Ultrasound-guided lumpectomy of nonpalpable
1996;83:1799. breast cancers: a feasibility study looking at the
17. Gordon PB, Gagnon FA, Lanzkowsky L. Solid accuracy of obtained margins. 1 Surg Onco11999;
breast masses diagnosed as fibroadenoma at 72:72-76.
fine-needle aspiration biopsy: acceptable rates of 31. Tafra L, Smith Sl, Woodward lE, et al. Pilot trial
growth at long-term follow-up. Radiology 2003; of cryoprobe assisted breast-conserving surgery
229:233-238. for small ultrasound visible cancers. Ann Surg
18. Drukker BH. Breast disease: a primer on diag- OncoI2003;1O(9):1018-1024.
nosis and management. Int 1 Fertil1997;42:278- 32. Staren ED, Sabel MS, Gianakakis LM, et al.
287. Cryosurgery of breast cancer. Arch Surg 1997;
19. Deschenes L, lacob S, Fabia 1, Christen A. 132:28-33.
Beware of breast fibroadenomas in middle-aged 33. Pfleiderer SO, Freesmeyer MG, Marx C, et al.
women. Can 1 Surg 1985;28:372-374. Cryotherapy of breast cancer under ultrasound
20. Kauffman CS, Bachman B, Littrup Pl, et al. guidance: initial results and limitations. Eur
Office-based ultrasound-guided cryoablation of RadioI2002;12:3009-3014.
breast fibroadenomas. Am 1 Surg 2002;184: 34. Sabel MS, Kaufman CS, Whitworth P, et al.
394-400. Cryoablation of early stage breast cancer: work
21. Larson TR, Robertson DW, Corica A, Bostwick in progress report of a multi-institutional trial.
DG. In vivo interstitial temperature mapping of Ann Surg OncoI2004;11(5):542-549.
35
Percutaneous Ablation of
Breast Tumors
Bruno D. Fornage and Beth S. Edeiken

The surgical management of breast cancer has logic examination of the margins is the only
evolved gradually over the past century from procedure that provides this crucial informa-
the exclusive use of radical mastectomy to the tion. Obviously, these ethical and technical lim-
current practice of segmental mastectomy and itations are of lesser concern in the treatment
radiation therapy. This less aggressive surgical of benign breast masses.
approach in the management of breast cancer Nonsurgical image-guided ablative tech-
extended to treatment of the axilla. The latter niques are available. Some of these techniques
led to the recent introduction and subsequent use devices that are inserted percutaneously
acceptance of lymphatic mapping and sentinel into the breast to either heat or cool the tumor
node biopsy as an alternative strategy to the sufficiently to cause complete cell death.
routine use of formal levels I and II axillary dis- Others, which involve the injection of chemical
section in clinically node-negative patients. agents such as pure ethanol, have been used in
In light of this trend toward less invasive the treatment of liver tumors, but these are
local therapy approaches for small tumors, increasingly losing popularity because they are
interest has emerged in evaluating nonsurgical difficult to control. Thus, the various modali-
ablative modalities for treating primary tumors. ties that are currently available to ablate
However, whereas previous applications of ra- breast lesions fall into two general categories:
diofrequency ablation (RFA) or cryotherapy- thermotherapy-hyperthermia induced by
such as RFA of bone metastases, RFA or application of radiofrequency current (i.e.,
cryotherapy of liver tumors, and cryotherapy of RFA) , laser irradiation, microwave irradia-
prostate cancer-often are performed for palli- tion, or insonation with high-intensity focused
ation and improvement of quality of life, not ultrasound (HIFU) waves-and cryoablation
cure, for patients with an otherwise poor prog- therapy.
nosis and for whom the therapeutic alternatives
incur more risk or simply are not available, the
challenge of minimally invasive therapy of
small breast cancer is that patients already have
Thermotherapy
an excellent prognosis. Therefore, if the percu-
taneous ablation fails, these patients may have
Radiofrequency Ablation
lost their best chance for cure of a clearly treat- With RFA, heat is produced through the appli-
able cancer. Besides this ethical issue, the main cation of a high-frequency alternating current
technical obstacle to the development of these that flows from the tip of an uninsulated elec-
therapeutic modalities is the inability to vali- trode into the surrounding tissue. The electrode
date the clear pathologic margins of the ablated itself is not the source of heat; rather, frictional
tissue. Currently, surgical excision with patho- heating is caused when the ions in the tissue

428
35. Percutaneous Ablation of Breast Tumors 429

attempt to follow the rapidly changing direc- solitary, and well visualized on sonography, and
tion of the alternating current. The tissue heats that its margins were well-demarcated from the
resistively in the area that is in contact with the adjacent tissues. We required the presence of a
electrode tip, and heat is then transferred con- l-cm distance between the tumor and both the
ductively to more distant tissue. skin and underlying chest wall for the lesion to
The promising clinical experience with this be eligible (Fig. 35.1). Before we performed the
modality in tumor sites other than the breast, RFA procedure, we took care to collect satis-
at the cost of minimal morbidity, provided factory core biopsy specimens to establish a
ample motivation to investigate its effective- definitive pathologic diagnosis, and to test the
ness in treating primary breast cancer. In tumor tissue for hormonal receptors and other
addition, the breast provides a favorable factors such as Her-2-Neu, because after RFA
environment because there are no major no other tumor tissue was available.
vessels that could cause convectional heat loss
(i.e., "heat sink") during the treatment, and
injuries to the skin or chest wall can be avoided
by excluding lesions that are too close to these
structures. On the negative side, primary breast
cancers are usually ill-defined, with irregular or
spiculated margins that are difficult to delineate
with sufficient accuracy by imaging. Only those
small invasive carcinomas that have clear-cut
margins and no or little associated ductal carci-
noma in situ (DCIS), determined by core
biopsy specimens, are good candidates for per-
cutaneous ablation.
The goal of RFA of primary breast cancer is
to achieve a lethally thermal lesion that encom-
passes not only the tumor, but also a margin of
surrounding normal tissue to destroy possible
peripheral microscopic disease. The initial A
report exploring the feasibility of RFA for the
treatment of breast tumors was published by
Jeffrey et al (1) on five women with locally
advanced invasive breast cancer, four of whom
had received preoperative chemotherapy with
or without radiation therapy. The authors con-
cluded that RFA was effective in causing inva-
sive breast cancer cell death, but that its use
would be applicable mostly to tumors smaller
than 3cm.
Radiofrequency ablation of the breast is
done under sonographic guidance and occa-
sionally under stereotactic guidance (2). In a
pilot "ablate and resect" study at the University
of Texas M. D. Anderson Cancer Center, we
investigated the feasibility of using sonographi- 8
cally guided RFA for the local treatment of FIGURE 35.1. Sonograms of a small mucinous carci-
small (Tl) invasive breast tumors (3,4). A noma of the breast. The tumor is well circumscribed
meticulous pre-RFA sonographic study was (arrow) and lies at a safe distance from both (A) the
performed to ensure that the tumor was small, skin surface (1.4cm) and (B) the chest wall (2.0cm).
430 B.D. Fomage and B.S. Edeiken

deployed over a distance of 3 em, and a radio-


frequency generator (Fig. 35.2). After we had
three-dimensionally located the target lesion,
the needle-electrode was inserted percuta-
neously through the tissues until its tip abutted
the lesion (Fig. 35.3). The prongs were then
deployed through the mass under full real-time
sonographic monitoring (Fig. 35.4). Then the
needle-electrode was connected to the genera-
tor, and the target temperature was set at 95°C.
Thermocouples at the tips of some of the
A prongs of the device provided continuous real-
time monitoring of the actual temperatures at
those sites. A laptop computer, using propri-
etary software developed by RITA Medical
Systems, displayed the curves of the tempera-
tures at these sites in real time, the power
output of the generator, and the impedance of
the tissues over time (Fig. 35.5). The target tem-
perature, once reached, was maintained for 15
minutes. No saline injections were used.
We noted no specific sonographic change
during the procedure that would reliably reflect
the pathologic changes induced by the RFA
and therefore help to determine the extent of
the thermal lesion. Instead, as the procedure
progressed, the sonographic visibility of the
lesion decreased, and sometimes the lesion was
obscured completely at the end of the proce-

B
FIGURE 35.2. Radiofrequency ablation instrumenta-
tion (RITA Medical Systems, Mountain View, CA).
(A) Photograph shows the multiarray needle-
electrode with its nine secondary prongs fully
deployed. (B) Photograph shows the radiofrequency
generator and the laptop computer, whose screen
graphically displays the temperatures recorded in
real time at the tips of five of the prongs.

For our study, we used a RITA RFA device


(RITA Medical Systems, Mountain View, CA), FIGURE 35.3. Sonographically guided insertion of the
which consists of a multiple-array needle- radiofrequency ablation needle-electrode. Photo-
electrode with several curved, flexible stainless graph shows that the cannula is aligned with the scan
steel secondary electrodes (prongs) that can be plane of the high-frequency linear-array transducer.
35. Percutaneous Ablation of Breast Tumors 431

A 8

FIGURE 35.4. Deployment of the prongs of the


radiofrequency ablation device. (A) Longitudinal
sonogram shows two prongs (arrows) traversing the
small carcinoma (arrowheads). (B) Transverse sono-
gram shows the cross sections of multiple prongs
(arrows) in the central portion of the rounded tumor
(arrowheads). (C) Diagram illustrates the expected
ablation volume achieved around the deployed
prongs of the multiarray device. c

dure by an ill-defined area of increased


echogenicity (Fig. 35.6). As we had expected,
color Doppler imaging performed after the
procedure was complete showed the complete
extinction of any preexisting vascularity in and
around the tumor within the ablated volume.
On gross pathologic examination, a hyper-
emic ring, measuring about 3 to 4 cm in diame-
ter, was seen around the tumor, representing
the outer limit of the ablated lesion (Fig. 35.7).
On hematoxylin and eosin staining, a thermal
effect was evident in the target lesions, includ-
ing the presence of disrupted cell outlines, pre- FIGURE 35.5. Screenshot of the laptop computer dis-
served nuclear staining, and increased plays the temperatures recorded at the tips of five of
cytoplasmic eosinophilia. Viability staining with the prongs, which have reached the target tempera-
reduced nicotinamide adenine dinucleotide ture of 95°C and have been maintained at that level
(NADH)-diaphorase confirmed the nonviabil- for 15 minutes (top). The graph also displays the
ity of the cells in the ablated volume. power output and the tissue impedance (bottom).
432 B.D. Fornage and B.S. Edeiken

c
FIGURE 35.6. Changes in the sonographic appear- the lesion (arrowheads). (C) Sonogram obtained at
ance of a small carcinoma during radiofrequency the end of the RFA session shows that the tumor is no
ablation (RFA). (A) Pre-RFA sonogram shows the longer identifiable owing to the substantial increase in
small (O.9cm), well-defined tumor (calipers). (B) echogenicity of the adjacent tissues, which obscures
Sonogram obtained at the beginning of the RFA pro- the lesion.
cedure shows the prongs (arrows) inserted through

FIGURE 35.7. Photograph of the breast tissue speci-


men excised after completion of radiofrequency
ablation (RFA) shows a reddish hyperemic ring
(arrows) defining the extent of the ablation zone.
Note the small tumor (arrowheads) in the center of
the ring and the tract of the RFA device through the
small tumor.
35. Percutaneous Ablation of Breast Tumors 433

No complications, such as burn of the over- risk of local recurrence, even if RFA is followed
lying skin or underlying chest wall, occurred, by radiation therapy. For this reason, ill-defined
probably as a result of our stringent patient tumors that are not well demarcated from sur-
selection criteria. A case of full skin burn during rounding tissues on imaging, including invasive
breast RFA has been reported in another study lobular carcinomas, should not be considered
(5). targets for curative RFA. Tumors with a sub-
In our study, the target lesion delineated by stantial DCIS component on diagnostic core
sonography was ablated completely in 100% biopsy specimens also should be excluded.
of the cases (Fig. 35.7). In one case of a tumor The use of real-time sonography during RFA
that had been downstaged by preoperative has been ineffective in both monitoring the for-
chemotherapy from a 4-cm palpable mass to a mation and assessing the extent of the thermal
sonographically and mammographically deter- lesions. By sonography, the margins of the
mined residua of about 1cm in diameter, RFA tumor are obscured by a diffuse area of hyper-
ablated the small residua; however, pathologic echogenicity and sometimes disappear com-
examination revealed extensive invasive and in pletely as the RFA progresses. Therefore, the
situ carcinoma around it. Therefore, we believe only-but critical-role of sonographic guid-
that patients who have been treated with pre- ance is to ensure that the device is placed in the
operative chemotherapy should not be offered exact geometric center of the target, and that
RFA as a therapeutic option. Our study has the prongs are deployed in such a manner as to
been expanded to include two other centers generate an ablation volume that will concen-
with less favorable results, raising the issues of trically encompass the tumor. For the time
both appropriate selection of cases and opera- being, until an imaging modality is developed
tor dependence. Thus, before RFA can be used that can accurately ascertain the margins of
as a replacement for the standard-of-care the ablation volume during the procedure, the
therapy of small breast cancer, that is, lumpec- use of RFA as a curative option for early
tomy followed by radiation therapy, important breast cancer requires a certain amount of
questions and serious concerns need to be overtreatmen t.
addressed. Will all ablated tumors proceed to complete
The optimal RFA treatment parameters, that necrosis? To date, we have information on the
is, those that ensure ablation of the tumor with effectiveness of the ablation derived only from
sufficient margin and the least damage to the immediate evaluation of the target tissue
surrounding tissues, are not yet fully estab- through special viability staining. Whether this
lished. They will likely vary, according to the type of assessment is predictive of eventual
histopathologic type of the tumor and the com- complete necrosis of the treated tumor if left in
position of the surrounding breast tissue. It is situ after RFA will need to be confirmed.
conceivable that our RFA protocol overtreats How difficult will it be after RFA to follow
the lesions that might be destroyed equally well patients with only standard imaging modalities
with a shorter RF session, lower target temper- (mammography and sonography) and physical
ature, or both. The use of animal models should examination? Whether extensive fat necrosis
provide information about this possible dose- and its associated early calcifications will inter-
effect relationship. fere with the early detection of recurrence
The true microscopic extent of the tumor, remains to be determined. Imaging with mag-
which may be very large and at times extremely netic resonance imaging (MRI), positron emis-
asymmetric, cannot be determined accurately sion tomography (PET), or both is expected to
either sonographically or by any other cur- play a crucial role in the follow-up of these
rently available imaging technique. In the patients. However, even the use of sophisticated
absence of surgical resection with pathologic imaging modalities may not prevent the need
evaluation of the specimen, any residual disease for extensive sampling of the periphery of
in or at the periphery of the ablated volume the ablated lesion with core needle biopsies to
would go undetected and could lead to a higher detect microscopic recurrent or residual disease.
434 B.D. Fornage and B.S. Edeiken

What will the long-term cosmetic results of operator needs to hit any part of the target
RFA be? Although it may be predicted that the lesion (including its edge) only once in four to
cosmetic results would be excellent, the extent six attempts. However, to ensure successful
of tissue retraction and fibrosis, and the ulti- RFA, the device must be placed in the geomet-
mate effect of radiation therapy on a thermally ric center of the tumor, which requires accurate
treated area not only are unknown, but may 3D mental images of the location of the tips of
vary from patient to patient. the deployed prongs and the anticipated shape
The most important end point for future and size of the ablation volume, with no room
trials designed to use RFA as an alternative for guesswork. Thus, the placement of the RFA
curative procedure to lumpectomy followed by device inside the target lesion requires a
radiation therapy, is long-term local control. greater level of skill than does merely hitting
Will RFA, which causes a coagulative necrosis the target with an automated biopsy gun. It is
of the tumor and a surrounding rim of tissue, doubtful that the expertise required to perform
adequately mimic a surgical procedure and curative RFA of breast cancer in terms of the
yield similar results in long-term local control? accuracy of the 3D placement of the device
Since the expected 5-year recurrence rate is and prediction of an acceptable size thermal
only a few percent with surgery, experience lesion is exportable to the general surgical
with large numbers of patients will be needed
to determine whether any statistically signi- Patients with breast tumor So 1.5 em
ficant difference exists between the two
treatments.
After our pilot RFA study at the University •
Informed consent


of Texas M. D. Anderson Cancer Center was
successful, we designed a protocol for the use
of RFA as a replacement for lumpectomy of Radiofrequency ablation of primary tumor'

cancers up to 1.5cm (Fig. 35.8) (6). Patients will .. 2- 4 weeks

undergo core biopsy at the periphery of the Core biopsy and fine-needle aspiration biopsy'
ablated zone 4 weeks after the RFA procedure
to assess local tumor control, and will undergo
imaging procedures and quality-of-life evalua-
tions at 6-month intervals for the first 2 years Biopsy negative
for residual tumor
Biopsy positive
for residual tumor
and annually thereafter through year 5. The
results of this study will indicate whether RFA
is oncologically and cosmetically appropriate
Radiation therapy Surgical resection
for the local treatment of primary breast +/- chemotherapy (Iumpectomy or mastectomy)
cancer. +/- hormonal therapy

~
The best results in all sonographically guided
interventional procedures are achieved by
radiologists highly skilled in sonography, and
Imaging studies to assess local control
1
Off study
the RFA procedure is no exception. RFA and change in RFA lesion size;
quality-of-life studies to assess
requires the highest level of eye-hand coor- functionality and cosmesis:
dination and three-dimensional (3D) sono- 'Years 1 & 2: Every 6 months
·Years 3 to 5: Annually
graphic "vision." A major issue regarding
the skills required for the procedure will be the • Assessment of axillary lymph node status may take place at either point,
degree of operator dependence. Although the at the discretion of the surgeon.

sonographically guided percutaneous insertion FIGURE 35.8. Protocol of a future study, designed
of the RFA probe appears similar to that of a to test radiofrequency ablation as an alternative to
core biopsy needle, there is a notable difference breast conservation surgery and radiation therapy
between the two procedures. For a sonographi- for the local treatment of small (:::;1.5 cm) breast
cally guided core biopsy to be successful, the carCInomas.
35. Percutaneous Ablation of Breast Tumors 435

community. We therefore believe that a team four metallic markers were placed in the tumor,
approach involving an expert interventional and one thermocouple was inserted at a dis-
sonologist is a prerequisite. tance of 1 cm from the laser fiber tip. Then 50
Which patients should be offered this alter- mL of local anesthetic were infused. The target
native therapy for breast cancer? Until RFA temperature, as measured by the thermocouple,
proves equivalent to breast conservation was 60°C. The average treatment time was 30
surgery and applicable to a wider range of minutes. The ablated lesions were surgically
patients, patients who are not candidates for removed for pathologic evaluation 1 to 8 weeks
surgery (or are poor candidates) would benefit later, and a 2.5- to 3.5-cm hemorrhagic ring was
from curative RFA, as would elderly patients seen surrounding the necrotic tumors. Com-
with slow-growing tumors and a lower risk of plete tumor ablation was achieved in only 70%
recurrence. Selected patients with local recur- of cases, but this figure includes both the results
rences also could be considered for RFA. obtained during the authors' learning curve and
Finally, some consideration should be given to technical failures. In two patients who refused
the concept of performing RFA (or any other to undergo subsequent surgical excision, the
percutaneous ablation technique) on a tumor unresected tumors first showed shrinkage, fol-
prior to its surgical excision in an attempt to lowed by the development of a 2- to 3-cm oil
minimize dissemination of tumor cells during cyst and fibrosis, proven by needle core biopsies
tumor manipulation. (9). This group of investigators also reported
earlier that they observed total tumor ablation
with negative margins whenever 2500J/mL of
Laser Irradiation tumor were administered or when the thermal
Laser-irradiation thermotherapy is another sensors recorded 60°C. In that study, micro-
ablation technique based on interstitial hyper- scopic examination 1 week after the ablation
thermia (7). Various lasers have been used for showed disintegration of malignant cells, with a
this purpose. Recently, diode lasers have largely peripheral acute inflammatory response and
replaced neodymium:yttrium-aluminum-garnet extensive fibrosis, noted 4 to 8 weeks after the
(Nd:YAG) lasers because of their small size, ablation (10).
portability, and lower cost. Laser ablation also can be performed under
An advantage of laser fibers is their small MRI guidance. An important advantage of
size: a bare fiber can be inserted through a MRI guidance is the availability of accurate
needle as small as 22 gauge. A treatment zone phase-sensitive temperature mapping that can
of about 1 em can be expected to result from a monitor the extent of the thermal lesion during
precharred laser tip when tissue is exposed to 2 the procedure (13).
to 3 W of continuous laser power for about 10
minutes. The laser energy may be split into as
Insonation with High-Intensity
many as four fibers for simultaneous treatment
Focused Ultrasound (HIFU) Waves
through multiple needles. Recently developed
diffuser tips have extended the treatment zone Another approach to local tissue destruction
to 3 em. Laser-irradiation thermotherapy has has been through the use of HIFU waves, deliv-
been performed under sonographic (8), stereo- ered by a series of transducers that focus the
tactic (9,10), or MRI (11,12) guidance. waves onto a predetermined area within the
In a recent study of stereotactically guided breast that rapidly heats to the point of tissue
laser ablation of breast cancer, 54 tumors (50 destruction. Unlike other ablation techniques,
invasive and four in situ; 51 masses and three insonation with HIFU waves does not involve
areas of microcalcification) with a mean diam- percutaneous insertion of a device inside the
eter of 1.2cm were treated on a stereotactic lesion. A disadvantage of this technique,
table, using a 16- to 18-gauge 805-nm laser however, is the substantial risk that the target
probe, with an optic fiber transmitting a pre- lesion will move during the procedure and
determined amount of laser energy (9). First, therefore be missed. Another limitation is the
436 B.D. Fornage and B.S. Edeiken

relatively small amount of tissue that can be damage, which is sufficient for complete cell
treated at a time. destruction at a final temperature of -40°C if a
Magnetic resonance imaging has been the 25°C/minute cooling rate is used and at -20°C
imaging modality of choice for guidance with if a 50°C/minute cooling rate is used (17).
the HIFU ablation technique. A few clinical In a study in rats, mice, dogs, and sheep that
research studies have been undertaken with used a 3-mm cryoprobe and liquid nitrogen for
HIFU of breast tumors, mostly concentrating cryoablation of mammary adenocarcinomas, a
on the treatment of fibroadenomas (14,15). single short «7 minutes) freeze killed only
In one series of 11 fibroadenomas in nine tumors smaller than 1.5 cm in diameter, despite
patients, HIFU waves were used under MRI an apparent temperature decrease to -40°C at
guidance with temperature-sensitive phase the periphery of the tumors (18). If the iceball
difference-based imaging during each sonica- fully encompassed the tumor and was main-
tion to monitor both the location of the focus tained for at least 15 minutes, 100% tumor kill
and the increase in temperature, which ranged was achieved, independent of tumor size. In this
from 13° to 50°C (15). Three of the 11 lesions study, two freeze-thaw cycles, each consisting of
were not treated adequately because of insuf- a 15-minute freeze and a lO-minute thaw, were
ficient acoustic power, patient movement, most effective. No procedure-related complica-
or both. One case of transient edema of the tions were noted (18). This same group of in-
pectoralis muscle was noted 2 days after the vestigators also treated one human patient
procedure (15). who had two invasive lobular carcinomas: the
specimens obtained during core needle bio-
psies performed 4 and 12 weeks after the well-
Cryoablation Therapy tolerated cryoablation procedure were nega-
tive for residual disease (18).
Cryoablation, once a popular technique for A more recent study involved 16 breast
treating liver metastases, is regaining the favor cancers with a mean (± standard deviation) size
of some investigators, especially for treating of 2.1 ± 0.8cm that were treated with a 3-mm
fibroadenomas. This is due in part to the advent cryoprobe inserted under sonographic guid-
of new, compact, easy-to-use cryoablation sys- ance for two freeze-thaw cycles of a 7- to 10-
tems that use argon gas, which reaches freezing minute freeze and a 5-minute thaw (19). The
temperatures faster than liquid nitrogen. mean diameter of the iceball after the second
Because the very cold temperatures achieved in freeze cycle was 2.8 ± 0.3 cm. Five tumors
cryoablation are anesthetic, this procedure can smaller than 1.6 cm showed no residual invasive
be performed safely under local anesthesia with cancer after treatment, but two of them had
little or no sedation of the patient. DCIS in the surrounding tissues, whereas
During cryoablation of both normal and tumors of 2.3 cm or larger showed incomplete
malignant breast tissue, ice initially forms in necrosis.
the connective tissue, then propagates and Cryoablation is well suited for use under
surrounds fat cells in normal adipose tissue sonographic guidance because the hyperechoic
and clumps of tightly packed malignant cells in front edge of the iceball that is created appears
cancer (16). sharp sonographically (Fig. 35.9). This allows
The thermal variables involved in cryoabla- accurate real-time visibility and control of the
tion include cooling rate, temperature gradi- extent of the thermal lesion, which is not pos-
ent, freezing interface velocity, final freezing sible with RFA. As a result, the procedure is
temperature, holding time at the final tempera- easily tailored to the size of the lesion. If
ture, warming (thaw) rate, and number of needed to protect the skin, sterile saline can be
freeze-thaw cycles (17). Cellular damage injected under sonographic guidance at the
increases with faster cooling because of the periphery of the iceball.
higher probability of intracellular ice forma- At our institution, we have used an argon-
tion. A double freeze-thaw cycle increases cell based device (Visica System; Sanarus Medical,
35. Percutaneous Ablation of Breast Tumors 437

A B

c o

E F
FIGURE 35.9. Cryoablation of a large fibroadenoma (E) Longitudinal sonogram obtained during the
in a 25-year-old woman who had undergone five cryoablation shows the iceball (arrows) encompass-
previous excisional biopsies for fibroadenomas. (A) ing the fibroadenoma. (F) Sonogram shows that
Sonogram shows a large fibroadenoma (calipers). saline is injected between the icebaB (arrowheads)
(B) Photograph shows insertion of the cryoprobe and the skin under sonographic guidance to avoid
through the breast under sonographic guidance. the development of skin lesions. Arrows point to the
(C) Longitudinal sonogram shows the cryoprobe needle. Note the marked reverberations behind the
(arrows) advancing through the center of the oval probe. (G) Photograph of the breast 24 hours later
lesion. (D) Transverse sonogram confirms the correct shows no evidence of hematoma. (H) Sonogram
position of the cryoprobe (arrow) through the obtained 8 months later shows a 75% reduction in
central portion of the target lesion. Note the typical the volume of the lesion.
comet-tail artifact associated with the metallic probe. (continued)
438 B.D. Fornage and B.S. Edeiken

G H
FIGURE 35.9. (continued)

Inc., Pleasanton, CA) with a cryoprobe that is fibroadenoma of less than 1.0cm, to 10 minutes
2.4 mm in diameter and is placed under sono- for a fibroadenoma of 3.6 to 4.0cm (Table 35.1).
graphic guidance. Two 8- to lO-minute cycles of Common transient postprocedure side ef-
freezing separated by a thaw of the same dura- fects of cryoablation of breast masses are local
tion usually have been applied to 2- to 3-cm swelling and ecchymosis, which resolve within 2
fibroadenomas. An improved algorithm that to 3 weeks. Treated fibroadenomas shrink over
allows more rapid treatment combines a cycle 6 to 12 months. In one multicenter study, 57
of active "high freeze" (to reach the target tem- core biopsy-proven fibroadenomas ranging
perature) and one of "low freeze" (to maintain from 0.8 to 4.2cm, with a mean size of 2.1cm,
the low target temperature). The respective were cryoablated using two freeze cycles for a
durations of the two cycles are tailored to the total duration of 6 to 30 minutes (20). The pro-
size of the lesion, and the total freeze time does cedures were performed under local anesthe-
not exceed 10 minutes; the relative duration of sia, most of them in an office setting. The
high freeze increases from 2 minutes for a median tumor volume decreased by 65 % at 6
months and 92% at 12 months. No skin injury
and little or no pain during and after the pro-
cedure were noted, and the cosmetic results and
TABLE 35.1. Relative durations of high and low
freeze cycles* for percutaneous cryoablation of fibro-
degree of patient satisfaction were excellent.
adenomas according to lesion size. The authors recommended the use of cryo-
ablation as an alternative to surgical excision of
Freeze cycle duration
(minutes) fibroadenomas (20).
Fibroadenoma
size (em) High Low
<1.0 2 o Conclusion
1.0-1.5 2 4
1.6-2.0 3 5
2.1-2.5 5 5
As more small, nonpalpable solid masses are
2.6-3.0 6 4 detected by screening mammography, the
3.1-3.5 8 2 natural desire in the medical community is for
3.6-4.0 10 o the development of systems capable of com-
* Freeze cycles are categorized as "high", for reaching the pletely destroying these masses in the least
target temperature, and "low", for maintaining the low invasive manner (i.e., with the least trauma,
target temperature. pain, and inconvenience to the patient), with
35. Percutaneous Ablation of Breast Tumors 439

optimal cosmetic results, yet without sacrificing 7. Steger AC, Lees WR, Walmsley K, Brown SG.
the chance for cure. Although several modali- Interstitial laser hyperthermia: a new approach
ties already appear capable of achieving this to local destruction of tumours. BMJ 1989;299:
goal, important issues remain to be resolved 362-365.
before they can become the standard of care for 8. Basu S, Ravi B, Kant R. Interstitial laser hyper-
thermia, a new method in the management of
breast diseases, particularly breast cancer. From
fibroadenoma of the breast: a pilot study. Lasers
a technical standpoint, a substantial amount of Surg Med 1999;25:148-152.
information on treatment methods remains to 9. Dowlatshahi K, Francescatti DS, Bloom KJ.
be collected for each percutaneous ablation Laser therapy for small breast cancers. Am J
technique ("How would you like your tumor- Surg 2002;184:359-363.
cooked or frozen?"). Eventually, in addition to 10. Dowlatshahi K, Fan M, Gould VE, Bloom KJ,Ali
being compared with current standards of care, A. Stereotactically guided laser therapy of occult
all these different techniques will need to be breast tumors: work-in-progress report. Arch
compared with one another ("Does frying kill Surg 2000;135:1345-1352.
better than freezing?"). Further studies, espe- 11. Harms SE. MR-guided minimally invasive pro-
cially ones evaluating long-term outcome, are cedures. Magn Reson Imaging Clin N Am 2001;
9:381-392.
mandatory before the current standard-of-care
12. Vogi TJ, Mack MG, Straub R, et al. MR-guided
treatment of small breast cancer can be altered. laser-induced thermotherapy with a cooled
Progress should be made only with caution and power laser system: a case report of a patient
without being driven by technology. Nonethe- with a recurrent carcinoid metastasis in the
less, these new treatment options are attractive breast. Eur Radiol 2002;12(suppl 4):S101-
alternatives, especially for benign breast dis- S104.
eases such as fibroadenomas, provided they 13. Huber PE, Jenne JW, Rastert R, et al. A new
prove to be both safe and efficacious. noninvasive approach in breast cancer therapy
using magnetic resonance imaging-guided
focused ultrasound surgery. Cancer Res 2001;61:
8441-8447.
References 14. Hynynen K, Pomeroy 0, Smith DN, et al. MR
imaging-guided focused ultrasound surgery of
1. Jeffrey SS, Birdwell RL, Ikeda DM, et al. Radio- fibroadenomas in the breast: a feasibility study.
frequency ablation of breast cancer: first report Radiology 2001;219:176-185.
of an emerging technology. Arch Surg 1999;134: 15. Hong JS, Rubinsky B. Patterns of ice formation
1064-1068. in normal and malignant breast tissue. Cryobiol-
2. Elliott RL, Rice PB, Suits JA, Ostrowe AJ, Head ogy 1994;31:109-120.
JE Radiofrequency ablation of a stereotactically 16. Rui J, Tatsutani KN, Dahiya R, Rubinsky B.
localized nonpalpable breast carcinoma. Am Effect of thermal variables on human breast
Surg 2002;68:1-5. cancer in cryosurgery. Breast Cancer Res Treat
3. Mirza AN, Fornage BD, Sneige N, et al. Radio- 1999;53:185-192.
frequency ablation of solid tumors. Cancer J 17. Staren ED, Sabel MS, Gianakakis LM, et al.
2001;7:95-102. Cryosurgery of breast cancer. Arch Surg 1997;
4. Fornage BD, Sneige N, Ross M, Singletary SE, 132:28-34.
Mirza AN, Edeiken BS. US-guided percutaneous 18. Pfleiderer SO, Freesmeyer MG, Marx C, Kuhne-
radiofrequency ablation in the local treatment of Heid R, Schneider A, Kaiser WA. Cryotherapy
small invasive breast carcinoma. Radiology 2002; of breast cancer under ultrasound guidance:
225(P):459. initial results and limitations. Eur Radiol 2002;
5. Izzo F, Thomas R, Delrio P, et al. Radiofrequency 12:3009-3014.
ablation in patients with primary breast carci- 19. Kaufman CS, Bachman B, Littrup PJ, et al.
noma: a pilot study in 26 patients. Cancer 2001; Office-based ultrasound-guided cryoablation of
92:2036-2044. breast fibroadenomas. Am J Surg 2002;184:394-
6. Singletary SE, Fornage BD, Sneige N, et al. 400.
Radiofrequency ablation of early-stage invasive 20. Kaufman CS, Bachman B, Littrup PJ, et al. Office
breast tumors: an overview. Cancer J 2002;8:177- based ultrasound-quided cryoablation of breast
180. fibroadenomas. Am J Surg 2002;184:394-400.
36
Complications of Tumor Ablation
Lawrence Cheung, Tito Livraghi, Luigi Solbiati, Gerald D. Dodd III,
and Eric vanSonnenberg

Percutaneous and surgical ablation procedures and equipment. Postprocedure follow-up with
are flourishing, in large part because of the rel- appropriate clinical, laboratory, and imaging
ative paucity and the acceptability of complica- studies is critical to detect and remedy any com-
tions. However, serious and fatal complications plications as early as possible.
have occurred, albeit rarely, with ablation. Complications of ablation should not be con-
This chapter identifies those complications, fused with those of the associated procedures
describes their underlying cause, and, when that may precede or follow the ablation, such
possible, discusses strategies to avoid them. as biopsy, fluid infusion techniques, or possibly
When initially considering a patient for surgery. Even fine-needle biopsy that may
tumor ablation, awareness of both the fre- immediately precede the ablation has its asso-
quency and scope of complications that can ciated complications that may be serious and
occur with the various procedures is necessary major (1).
to accurately weigh the potential benefits of the In classifying complications as major or
intervention against its attendant risks. These minor, a major complication is one that, if left
risks also should be evaluated relative to those untreated, might threaten the patient's life, lead
of the alternatives under consideration, such as to substantial morbidity and disability, or result
surgical resection, aggressive chemotherapy, or in a lengthened hospital stay. All other compli-
a course of radiation therapy. cations are considered minor. They will be cat-
Although each local ablation technique pos- egorized in this chapter primarily according to
sesses its own characteristic dangers, there are the mechanism of ablation, and by the organ in
general guidelines that can be followed to min- which they occur. We include the reported lit-
imize the severity and rate of complications. erature, as well as our own personal experience.
Preprocedure, patients should be evaluated
carefully with regard to lesion size and location,
feasibility of access routes, and degree of under- Percutaneous Ethanol
lying clinical comorbidities. Bleeding and
infection are potentially common to all inter-
Injection (PEl)
ventional procedures; thus, coagulation tests
should be performed on all patients and nor-
Hepatic
malized whenever possible, and prophylactic Percutaneous ethanol injection most frequently
antibiotics ought to be considered for those is used for liver tumors, primarily hepatocellu-
at increased risk of infection. The procedure lar carcinoma (HCC). In numerous large
itself should be performed meticulously, with studies of PEl in the liver, it has consistently
attentive monitoring of vital signs, and care been shown to be a safe procedure, with mor-
given to maintaining the sterility of the field tality rates ranging from 0.09% to 0.97%, and

440
36. Complications of Tumor Ablation 441

major complication rates of 1.7% to 4.6% hepatic necrosis (4,6). A case complicated by
(2-4). Some of these studies include data for hemothorax was performed using an intercostal
both conventional and single-session PEl. Con- approach and was thought to be due to acci-
ventional PEl generally is used to treat lesions dental rupture of the intercostal artery (4). A
less than 5 cm in greatest dimension, and case of hepatic necrosis was attribute.d to an
involves injecting small amounts of ethanol, intratumoral arterioportal shunt that allowed
ranging from 1 to 15 mL depending on lesion the injected ethanol to spread diffusely
size. It does not require general anesthesia and throughout the liver (6).
often is performed in multiple sessions. In con-
trast, single-session PEl is used to treat large Major Complications
lesions (>5cm) by performing multiple passes
Major complications that can occur with PEl in
and injecting significantly more ethanol during
the liver include variceal bleeding, intraperi-
a single session. The procedure is performed
toneal bleeding, and hemothorax as discussed
under general anesthesia, and the volume of
above, as well as hemobilia, subcapsular hema-
ethanol injected can exceed 100mL. If the
toma, portal vein thrombosis, biloma, sepsis,
results of conventional and single-session PEl
liver abscess, septic cholangitis, hepatic infarct
are analyzed separately, single-session PEl has
of segments adjacent to the lesion, acute liver
increased complication rates, with one study
failure, and worsening of hepatic function with
reporting mortality and major complication
Child-Pugh class progression. The presence of
rates of 1.71 % and 6.29%, respectively, for
a biloenteric anastomosis is a major risk factor
single-session PEl, compared with 0.27% and
for septic complications either during or soon
0.54% for conventional PEl (4).
after PEl (7), and postprocedure fevers in these
patients should be evaluated carefully. Prophy-
Mortality lactic antibiotics are mandatory for ablation in
these patients.
Reported causes of death from PEl in the liver
Another major complication of PEl is neo-
include bleeding gastroesophageal varices,
plastic seeding along the needle tract, with a
hemoperitoneum, and acute liver failure (2-5).
reported incidence ranging from 0.01 % to
Bleeding varices are the result of worsening
1.4%. Tumor seeding may be seen intraperi-
portal hypertension from the procedure. When
toneally in the liver and abdominal cavity, sub-
ethanol is injected into a lesion, it causes throm-
cutaneously in the chest wall or abdominal wall,
bosis of tumor vessels; however, it also can
along the diaphragm, and cutaneously (8-10).
induce thrombosis of small vessels in regions of
Metastatic tumor spread to the lung also has
the liver adjacent to the lesion. This leads to a
occurred following PEl in the liver (11),
sudden increase in venous portal pressure, and
leading to the concern that partial tumor
if superimposed upon chronic portal hyperten-
necrosis following ethanol injection may in-
sion from advanced cirrhosis, can result in
crease embolization of tumor cells; however,
massive variceal bleeding.
there have been no other descriptions of this
Most instances of fatal intraperitoneal hem-
phenomenon.
orrhage are caused by the rupture of highly vas-
Other rare complications of PEl include
cular HCCs that are located superficially, that
syncope, hypotension, hypoventilation, pneu-
is, either against or protruding into the liver
mothorax, gastric ulcer formation, splenic
capsule (2,4). Death from acute liver failure
abscess, intestinal perforation, and acute renal
after PEl occurs primarily in patients with
failure from acute tubular necrosis (2-5,12-14).
Child-Pugh class B or C cirrhosis, as their
limited residual liver function renders the
Minor Complications
patient vulnerable to acute decompensation,
even from mild to moderate insults (4). The most common minor complications fol-
Other causes of death from PEl that have lowing PEl are pain, fever, nausea, and vomit-
been reported include hemothorax and massive ing, all of which are self-limited. Transient
442 L. Cheung et al

intoxication also is seen, especially with single- and relative immobility when compared with
session PEl, during which the large volume of the small bowel. Characteristic of thermally
injected ethanol can lead to blood alcohol induced perforation is having the target lesion
levels greater than 100mg/dL (5). Asympto- located within 1cm of the liver capsule and
matic right pleural effusions occasionally are adjacent to a gastrointestinal lumen. A second
detected on routine follow-up imaging, and risk factor for intestinal perforation appears to
generally resolve spontaneously. be prior abdominal surgery with the formation
of fibrotic adhesions that affix parts of the gas-
trointestinal tract to the liver (17). These adhe-
Thyroid sions probably impair normal peristalsis and
There has been limited usage of PEl to treat preclude movement of the bowel away from
benign thyroid nodules. Pain and fever are the the liver, thereby resulting in intraprocedural
most commonly reported side effects after heating.
the procedure. Other complications include Acute liver failure following RFA can be
transient dysphonia and Horner's syndrome secondary to various causes. There have been
(15,16), which relate to the proximity of the several cases of total portal vein thrombosis,
thyroid nodule to the recurrent laryngeal nerve from both intraoperative RFA with application
and cervical sympathetic chain, respectively. of the Pringle maneuver, and from percuta-
Common jugular vein thrombosis also has been neous RFA without balloon occlusion of the
reported as a minor complication that resolves portal vein (18). One patient developed liver
without treatment (16). failure secondary to right hepatic duct stenosis
following ablation of a lesion near the hepatic
hilum (17). Apparently, the large bile ducts are
Radiofrequency Ablation (RFA) susceptible to thermal damage; thus, lesions
near the hepatic hilum or large bile ducts
Hepatic should be approached with care. Another case
of fatal acute liver failure occurred in a patient
Mortality rates for hepatic RFA have ranged
who underwent right hepatectomy that was
from 0.3% to 1.6%, and major complication
extended to segment IV along with RFA of a
rates, from 2.4% to 5.7% (17,18). The risk of
lesion in segment III (18). A large area of
major complications increases with the number
radiofrequency destruction was found in the
of ablation sessions, although not with tumor
remaining liver, and the patient lacked suffi-
size (17). The choice of single versus cluster
cient residual function to compensate.
electrodes has not been found to affect compli-
Other deaths following hepatic RFA include
cation rates significantly (17). In one study that
a case of bile pulmonary embolism (19), one
compared percutaneous and intraoperative
patient with pancreatic cancer who died sud-
RFA, the mortality rate was 0.9% (2/216) for
denly 3 days postprocedure of unknown
percutaneous RFA, compared with 2.4%
cause, and a patient with obesity and diabetes
(3/123) for intraoperative RFA (18). Similar
mellitus who developed peritonitis from
types of complications, both major and minor,
Staphylococcus aureus that was attributed to
were seen in the two groups.
an unknown break in sterile technique (17).
Mortality
Major Complications
Causes of death following RFA in the liver
include intestinal perforation with subsequent In addition to the complications discussed
peritonitis and multiorgan failure, acute liver above, other major complications following
failure, and massive hemoperitoneum following hepatic RFA include liver abscess formation,
rupture of a superficial HCC nodule (17,18). tumor seeding, hypoxemia, and hemothorax.
The colon has the highest risk of perforation Patients with biloenteric anastomoses are at
from thermal damage, likely due to its thin wall significantly higher risk for developing liver
36. Complications of Tumor Ablation 443

abscesses, similar to PEl (17,18). Patients with levels. This has prompted speculation that
diabetes mellitus also appear to be at higher intraprocedural heating of the adrenal gland
risk for septic complications (17). Patients with may stimulate the release of catecholamines.
either of these conditions should receive pro- Therefore, the possibility of a hypertensive
phylactic antibiotics prior to RFA. crisis should be considered when ablating liver
The incidence of tumor seeding following lesions near the right adrenal gland.
RFA in the liver is around 0.5% in most large
studies (17,18,20), although rates as high as Minor Complications
12.5% (4/32) have been reported in one study
The most common minor complication follow-
with a limited number of patients (21). The
ing RFA in the liver is postprocedure low-grade
risk of tumor seeding is increased with sub-
fever and general malaise, which has been
capsular lesions, highly undifferentiated HCC,
described as the "postablation syndrome" (27).
and high baseline serum a-fetoprotein level
Symptom severity and duration correlate
with HCC lesions (17,21). To decrease the risk
roughly with the volume of tumor ablated,
of tumor seeding, the initial probe placement
and more aggressive procedures tend to result
should be performed as carefully as possible
in more severe and prolonged symptoms.
to minimize the number of passes through
Although the symptoms generally resolve
the liver. Furthermore, after the ablation is
within 2 to 7 days, the syndrome can persist for
complete, RF energy can be delivered to the
up to 3 weeks with very large areas of necrosis.
probe during its withdrawal, thus cauterizing
Other minor complications include peripro-
the probe tract.
cedural and postprocedural pain, self-limiting
Hypoxemia has been seen in a small number
intraperitoneal and subcapsular bleeding, asymp-
of patients following RFA. The cause remains
tomatic arterioportal shunts, small pleural
to be elucidated, although there is speculation
effusions, and pneumothoraces that do not
that it is caused by acute lung inflammation
require treatment. Skin burns were seen much
from the overexpression of cytokines that has
more frequently before comprehensive guide-
been demonstrated with experimental cryoab-
lines for grounding pad safety were developed
lation in animals, but has yet to be seen with
(28).
RFA (22,23). Although the hypoxemia is tran-
sient and self-resolving after several days, it
may prolong hospital stay due to the need to Pulmonary
rule out pulmonary embolism.
The use of RFA to treat pulmonary lesions,
Major complications that have been reported
either primary or metastatic, is relatively new,
rarely include segmental hepatic infarction,
and data on the complications of this procedure
common bile duct stenosis, biloma, blood in the
are limited. Nevertheless, the initial reports on
gallbladder from hemobilia, acute cholecystitis,
pulmonary RFA, all of which have employed a
sepsis, hyperkalemia, diaphragmatic paresis,
percutaneous approach, indicate that it is a safe
pulmonary embolism, pleural effusion re-
procedure, with mortality rates ranging from
quiring thoracentesis, pneumothorax requiring
0% to 3.7% and major complication rates of
drainage, and cardiac arrest (17,18,24,25).
11 % to 30% (29-34).
There also has been a report of two cases
of severe intraprocedural hypertension requir-
Mortality
ing temporary cessation of the procedure and
the administration of intravenous labetalol or Reported deaths following pulmonary RFA
hydralazine (26). Both of these patients had include one case of acute respiratory distress
lesions in the posterior aspect of the right lobe syndrome (30) and two pulmonary hemor-
of the liver, in proximity to the right adrenal rhages (31,35). The case of acute respiratory
gland. In one patient, measurement of cate- distress syndrome occurred 4 days after the
cholamines during the hypertensive crisis RFA procedure in a patient who had recently
revealed massive elevations above normal been diagnosed with pneumonia. This has led
444 L. Cheung et al

to speculation that inflamed, consolidated lung require drainage, as high as 67% (four of six) in
parenchyma is more susceptible to thermal one small series (32).
damage from RF energy due to a difference in Common postprocedure complications in-
its conductivity relative to normal, aerated lung. clude fever and malaise, consistent with the
Until more data are available regarding this postablation syndrome, as well as pain and
complication, it is recommended that patients dyspnea. Other minor complications that have
with pneumonia should wait until the consoli- been reported are asymptomatic pleural effu-
dation has resolved completely before under- sion, mild intrapulmonary hemorrhage with
going RFA. transient hemoptysis, expectoration of small
Concerning the two pulmonary hemor- amounts of desiccated tissue (29), subcuta-
rhages, one was detected within 2 hours of the neous emphysema, and severe myalgia. From
procedure in a patient who was taking clopido- our own experience, we had one patient who
grel, an inhibitor of platelet aggregation, at developed a skin burn on the back, and another
the time of the procedure (35). Based on this with transient dysphonia following RFA of a
report, the authors advise that patients taking lesion in proximity to the aortic arch, near the
clopidogrel should discontinue the medication origin of the left recurrent laryngeal nerve.
1 week prior to RFA, similar to elective surgery
when bleeding is a risk. The other reported case
of pulmonary hemorrhage occurred 2 days Renal
after treatment and also was attributed to Various centers currently perform RFA to treat
platelet dysfunction (31). renal lesions; early experience indicates that
there is minimal morbidity and mortality asso-
Major Complications ciated with the procedure (36-40). With over
The primary major complication of pulmonary 100 patients reported, there has been only one
RFA is pneumothorax requiring thoracostomy death following renal RFA-a patient with a
tube placement for drainage. The incidence of history of severe pulmonary disease who died
pneumothorax that warrants drainage ranges from aspiration pneumonia 3 days after an
from 6.6% to 30% in the largest studies uneventful procedure (39). There was no evi-
(29-31). The risk of pneumothorax is signifi- dence of renal hemorrhage or direct injury to
cantly increased when the RF probe traverses the lungs from the ablation itself.
normal lung parenchyma, and pneumothorax
occurs more frequently in centrally located Major Complications
tumors than in those that are peripheral (30).
The few major complications reported are rep-
Whenever possible, a transparenchymal route
resentative of problems that have been seen
to the lesion should be avoided.
with RFA in the liver, such as bleeding, injury
Other reported major complications include
to adjacent structures, and tumor seeding. One
a cerebral infarction thought to be secondary to
patient with a central lesion developed a large
an air embolism (34), obstructive pneumonia
perirenal hematoma and blood in the renal
(30), and a large pleural effusion complicating
pelvis. A clot formed in the collecting system
a pneumothorax (29).
causing anuria, and the patient required cysto-
scopic ureteral stent placement and a blood
Minor Complications
transfusion (36).
Two intraprocedural complications that have Another patient suffered a small burn injury
been reported are persistent cough that re- to the liver immediately adjacent to a right-
quires a reduction in RF energy from >100 W sided, anterior renal lesion during RFA (39).
to 80W (30), and pain that necessitates a con- Injuries to adjacent structures are thought to
version from local anesthesia to intravenous be relatively rare, because of the surrounding
sedation (29). There is also a high incidence of perirenal fat. This provides a buffer that may
intraprocedural pneumothorax that does not help dissipate heat and protect nearby organs.
36. Complications of Tumor Ablation 445

A single instance of tumor seeding has been hematuria. Other reported minor complica-
reported following renal RFA. The patient tions include transient intraprocedural hyper-
developed a 5-mm, cutaneous metastasis at the tension (mean blood pressure, 215/100 mm Hg)
electrode insertion site, which was immediately that resolved spontaneously after the proce-
resected and has not recurred during the 16 dure (40), subcapsular hematoma (38), pain
months since the procedure (40). This compli- with hip flexion, and paresthesia in the perium-
cation illustrates the importance of careful clin- bilical region or the skin of the ipsilateral flank
ical follow-up. As in the liver, cauterization of (37,40). The cases of pain during hip flexion
the probe tract may help prevent tumor seed- resolved without treatment within 2 weeks,
ing if the probe has traversed normal renal and may reflect temporary injury to the psoas
parenchyma. muscle. The instances of paresthesia are likely
Another important question that has been due to injuries to the intercostal, lumbar, ilioin-
raised is whether the renal collecting system is guinal, or iliohypogastric nerves that are
susceptible to thermal injury during ablation of located close to the kidney. In some cases, the
centrally located lesions. Although a number of paresthesia persisted for months, and may indi-
deep, medial lesions have been ablated without cate permanent nerve damage.
complications (36,37), there are two recent
reports of injuries to the collecting system
(40,41). One patient was incidentally found to
Bone
have a dilated upper pole calyx on routine In a multicenter study of percutaneous RFA of
follow-up computed tomography (CT) scan bony metastases, 43 patients were treated, and
obtained 2 months after the procedure; this was one (2.3%) major complication was seen (42).
believed to be secondary to heat-induced stric- The patient suffered an acetabular fracture 6
ture of the collecting system (40). A more weeks after RFA of a lesion that involved the
serious case has been reported in a patient who acetabulum, ileum, and ischium. Although the
developed flank pain, mild hydronephrosis, and presence of tumor alone predisposes to patho-
mildly delayed contrast excretion 2 weeks after logic fracture, it is possible that tumor necrosis
treatment. Six months after ablation, the in the acetabulum from the ablation also con-
patient had persistent, severe hydronephrosis tributed. Two minor complications also were
and delayed contrast excretion; anatomic and reported in the study: one second-degree skin
functional imaging revealed complete uretero- burn at the grounding pad site and one case of
pelvic junction obstruction and that the transient bowel and bladder incontinence fol-
affected kidney was contributing only 8% to lowing RFA of a previously irradiated sacral
overall renal function. Open nephrectomy was metastasis.
performed and confirmed the presence of a In one of the only reported cases of intrauter-
fibrotic lesion in the collecting system at the site ine RFA during pregnancy, the ablation of a
of RFA (41). These studies suggest that fibrosis sacrococcygeal teratoma resulted in the birth of
and stricturing of the renal collecting system a newborn with an open posterior hip disloca-
with subsequent obstruction are potential com- tion and loss of sciatic nerve function (43).
plications of renal RFA, and that care should
be taken to avoid including the renal pelvis or
ureteropelvic junction within the treatment
Adrenal
zone whenever possible. In a series of 15 adrenal lesions in eight patients
treated with RFA, the only complication was a
delayed polymicrobial abscess in a 9-cm lesion
Minor Complications
that was discovered 11 weeks after the proce-
The most common minor complications fol- dure (44). The abscess was treated successfully
lowing renal RFA are periprocedural pain and with prolonged catheter drainage and antibi-
nausea, asymptomatic perirenal hematomas, otics. Another series of 13 adrenal masses in 12
and transient microscopic or macroscopic patients had no major complications (45). The
446 L. Cheung et al

only minor complication reported was a small Two retrospective studies have attempted to
hematoma in a patient with preexisting throm- compare the complication rates of cryoablation
bocytopenia from chemotherapy. No patients in and RFA for hepatic tumors using either an
either study developed hypertension during the open or percutaneous approach (50,59). In the
procedure, despite a recent report of cate- study that performed either cryoablation or
cholamine release and life-threatening hyper- RFA intraoperatively, the rate of major com-
tension during the ablation of liver lesions near plications for cryoablation was 40.7% (22/54)
the right adrenal gland (26). versus 3.3% (3/92) for RFA (59). However, the
other study that used a percutaneous approach
for both procedures failed to show significant
Breast differences in the major complication rates:
Several pilot studies have examined RFA of 29% (9/31) for cryoablation and 26% (8/31) for
breast lesions followed by surgical resection RFA (50). The choice of treatment modality
(46-49). The only major complication occurred was based on the preference and training of the
in a study of 26 patients, one of whom individual surgeons in the first study, and on the
developed a full-thickness skin burn following random availability of probes in the second. A
RFA of a lesion immediately beneath the skin third study that used the laparoscopic approach
(46). The minor complications reported were treated lesions smaller than 5 cm with RFA and
small breast ecchymoses in some patients, and those from 5 to 8 cm with cryoablation. The
there was one 5-mm skin burn that did not authors found major complication rates of 24%
require treatment (47,48). In one study of 20 (4/17) for cryoablation, and 3.4% (1/29) for
patients, there were no complications reported RFA (51).
(49).
Mortality
One of the most common causes of death
Cryoablation following hepatic cryoablation is a syndrome
consisting of thrombocytopenia, disseminated
Hepatic intravascular coagulation, acute respiratory dis-
Cryoablation of hepatic tumors has been used tress syndrome, hypotension, and multiorgan
since the 1980s, traditionally via laparotomy. failure. This constellation of symptoms has been
Currently, laparoscopic and percutaneous termed the "cryoshock phenomenon", due to its
approaches are gaining popularity (50-52). similarities to septic shock, although without
Mortality rates following open cryoablation evidence of systemic infection (60). Although
range from 1.2% to 7.5%, and major complica- some patients have the full syndrome, many
tion rates vary from 5.9% to as high as 45.0% have various combinations of the primary symp-
(53-56). Numerous studies have observed a toms. The exact mechanism of cryoshock
correlation between the volume of liver being remains to be elucidated; however, there is
ablated and the risk of major complications, evidence that large-volume freezing and
such as parenchymal fracture, coagulopathy, multiple freeze-thaw cycles stimulate release of
and acute renal failure from myoglobinuria cytokines similar to those seen in septic shock,
(55,57,58). The specific liver volume at which including tumor necrosis factor-a and
complications begin to increase significantly interleukin-6 (23,61). The cryoshock phenome-
has been estimated at >30% to 35% liver non is highly specific, both to cryoablation and
volume, lesions >5 to lOcm diameter, and a to the liver as the target organ. However, it is
total estimated area of lesions >30cm2 seen following cryoablation of both primary
(53-55,57). Currently, there is no evidence liver tumors and metastatic lesions, which sug-
regarding the relative safety of open, surgical gests it may be the surrounding, normal liver
cryoablation versus the laparoscopic or percu- tissue that is releasing the mediators of
taneous routes. cryoshock in response to cryotrauma, rather
36. Complications of Tumor Ablation 447

than the tumor. It is known that the Kupffer cells as mortality. In addition to these, the most
of the liver have a propensity to release common complications following cryoablation
cytokines following injury, including tumor are hypothermia, injury to the biliary system,
necrosis factor-a and interleukin-6. Another renal insufficiency secondary to myoglobinuria,
explanation for the specificity of cryoshock to hepatic abscess, and symptomatic pleural
the liver could be that the volume of tissue effusion.
frozen with hepatic cryoablation typically is Hypothermia associated with the cryoabla-
greater than with prostate or renal cryoablation. tion procedure can be severe, with a drop in
Severe hemorrhage is another major cause core temperature below 33.5°C. In addition to
of mortality associated with cryoablation. the cardiac complications of arrhythmia and
Although the coagulopathy of cryoshock may myocardial ischemia, hypothermia can induce
contribute to the high frequency of hemor- or worsen coagulopathy. The incidence of
rhage, the predominant source of significant hypothermia can be decreased by using Bair
bleeding comes from fracturing of the liver Hugger heating blankets (Arizant Healthcare,
parenchyma during the rapid freeze-thaw Prairie, MN) during the procedure and by
cycles, analogous to the cracks that form in an increasing the patient's core temperature to
ice cube when placed in warm water. The cracks 38.2°C prior to performing cryoablation
in the hepatic parenchyma extend from the (53,63).
center of the iceball to the outer margin of the Injuries to the biliary system are also poten-
freeze zone. In most instances, the bleeding tial complications. Although the large blood
comes from the fractured hepatic vessels, vessels are protected from thermal injury by the
although there is one report of a fatal hemor- warming effect of flowing blood, the large bile
rhage from the iceball cracking into the right ducts are susceptible to freezing damage.
hepatic vein (59). The risk of parenchymal Necrosis of the bile duct epithelium can lead to
fractures can be decreased by minimizing any bile leakage with biloma formation, biliary
movement or torque on the cryoprobe during strictures, or biliary-cutaneous fistulas. If the
the freezing process, especially when the freeze tumor to be ablated lies near the large bile
is near completion and the liver is at its coldest. ducts, one technique for protecting them from
Furthermore, attempts to retract the probe thermal damage is to circulate warm saline
should not be made until the iceball has thawed through a catheter placed in the bile ducts (64).
completely, thus allowing the probe to be Another technique that has been described
removed with minimal resistance. involves the placement of metal stents in the
Other causes of death following cryoablation large bile ducts approximately 2 days prior to
include liver failure from insufficient residual cryoablation (53). The stents help preserve the
function, colonic perforation from freezing integrity of the bile ducts during the freezing
injury, sepsis, acute myocardial infarction, and process, and may aid in their reepithelialization
pulmonary embolism. Rare causes of death if injured.
include variceal hemorrhage, hepatorenal syn- Myoglobinuria is seen in many patients fol-
drome, portal vein thrombosis, strangulated lowing cryoablation. Although usually tran-
bowel obstruction, acute pancreatitis, ischemic sient, myoglobinuria occasionally can progress
stroke, and delayed intraabdominal rupture of to acute tubular necrosis with renal insuffi-
the cryoablated area that led to multiorgan ciency. The incidence of acute tubular necrosis
failure (50,54,55,61,62). can be reduced, albeit not entirely negated, by
ensuring adequate hydration prior to the pro-
cedure, initiating mannitol diuresis during the
Major Complications
procedure, and alkalinzing the urine postproce-
Cryoshock, parenchymal fracture with subse- durally (53,65).
quent hemorrhage, freezing injury to adjacent Hepatic abscesses can develop in the ablated
structures such as the colon, and acute liver tumor, generally in the weeks following the
failure are all major causes of morbidity, as well procedure. Delayed abscess formation, more
448 L. Cheung et al

than 1 year after the procedure, also has been (56). Major complication rates are discussed
observed (66). Many patients develop pleural individually for the most common problems,
effusions following hepatic cryoablation, and and although many of these, such as impotence
while most are asymptomatic and resolve in less and incontinence, are not life threatening, they
than 2 weeks, those that cause dyspnea or do do have a significant impact on quality of life.
not resolve spontaneously require thoracente- Salvage cryoablation following failed radio-
sis or placement of a thoracostomy tube. therapy has higher complication rates than
Other reported complications include sub- primary cryoablation (74), as is discussed
capsular hematoma, wound infection, wound further below.
dehiscence, injury to the diaphragm, pneu-
mothorax, right lower lobe pneumonia, mental
Major Complications
status changes, unexplained fever, severe
nausea and vomiting, and acute pancreatitis Impotence is the most common complication
(51,54,59,61). following prostate cryoablation, with reported
rates of 53% to 93% in previously potent indi-
Minor Complications viduals (71-73,75). The neurovascular bundles
in the prostate that carry the cavernosal nerves
Minor complications following hepatic cryoab- responsible for erectile function generally are
lation include the postablation syndrome of ablated during the procedure to ensure com-
low-grade fever and malaise, periprocedural plete destruction of the tissue at the periphery
and postprocedural pain, nitrogen skin burns of the prostate gland. It is hypothesized that
(51), transient elevation of liver enzymes, since the nerves are not cut, they have the
asymptomatic pleural effusions, myoglobinuria, potential to recover their function over time,
and transient thrombocytopenia or coagulopa- and that patients may recover potency. One
thy that does not require transfusion of study reported that 19 of 373 patients (5.1 %)
platelets or fresh frozen plasma. regained erectile function over an average of
16.4 months (73), while another reported that at
3 years, 18 of 38 (47%) had resumed sexual
Prostate intercourse, five (13 %) spontaneously and 13
Although prostate cryoablation first began to (34%) with the aid of sildenafil or prostaglandin
be performed in the late 1960s (67), complica- (75). Nerve-sparing, unilateral cryoablation cur-
tion rates were unacceptably high, due to the rently is being investigated, and in a pilot study,
inability to place cryoprobes precisely or to seven of nine patients (78%) retained potency
monitor the freezing process accurately. Tech- following the procedure (76).
nological advances in the past 15 years, which Urinary incontinence is another common
include transrectal ultrasound guidance and complication, although the exact incidence
monitoring (68), as well as placement of a ure- varies both with the method of assessment and
thral warming device (69), markedly decreased whether the patient is undergoing primary or
complications. A recent development is the salvage cryoablation. Reported incontinence
transition from liquid nitrogen to gas-driven rates for primary cryoablation have ranged
probes that permit the use of cryoprobes with from 7.5% to 15.9% using a broad definition
a much smaller diameter; this avoids the need that includes any degree of leakage (71-73).
for tract dilatation, and allows the direct place- The incidence of severe incontinence that
ment of more probes into the tumor (70). requires the daily use of urinary pads has been
Mortality following prostate cryoablation is reported from 3% to 4.3% (71,73). Early
extremely rare, with most large studies report- studies that examined salvage cryoablation fol-
ing no deaths (71-73). A multicenter survey lowing failed radiotherapy found postproce-
reported only three deaths out of 5432 patients dure incontinence rates as high as 73% (74),
(0.06%), two from pulmonary embolism, and while in more recent studies only 7.9% to 20%
one from a complicated rectourethral fistula required urinary pads (77,78).
36. Complications of Tumor Ablation 449

Urethral sloughing occurs when the urethra tory medications once abscess and urinoma
is injured by the freezing process and allows have been ruled out. Osteitis pubis, or inflam-
necrotic prostate tissue to enter the urinary mation of the pubic symphysis, can be managed
tract. The signs and symptoms of urethral similarly. The scrotal swelling and penile
sloughing include dysuria, pyuria, and obstruc- paresthesia usually are transient and resolve
tion. Catheterization sometimes is successful spontaneously.
at dislodging the obstructing tissue, as well
as facilitating urinary drainage; however,
more severe cases may require transurethral Renal
resection of necrotic prostate tissue. The inci- A small but increasing number of centers have
dence of urethral sloughing that necessitates begun performing renal cryoablation via an
transurethral resection of the prostate ranges open (83-85), laparoscopic (85-88), or percuta-
from 2.5% to 13% in patients who undergo neous (89) approach. Although the total
primary cryoablation (72,73,79), with compara- number of patients treated is relatively small, it
ble results for salvage therapy (74,77). It is has proven to be a safe procedure with no
important to use an approved urethral warming reported deaths and relatively few major or
device, as these devices offer better protection minor complications.
against urethral sloughing than those that have
not been approved (72).
Major Complications
Rectourethral fistula formation is a severe
complication that can occur if the freeze zone An important major complication that has been
extends too far posteriorly. Although formerly reported is renal parenchymal or capsular frac-
a common problem prior to the implementa- ture during the thawing cycle that may lead to
tion of transrectal ultrasound monitoring, the hemorrhage (83,86). The technique to minimize
incidence of rectourethral fistula formation in the risk of renal fractures is similar to that in
recent large studies ranges from 0% to 0.5% for the liver: cryoprobes should be inserted gently
primary cryoablation (71-73). Rates are slightly with a minimum of torque, they should not be
higher in those undergoing salvage therapy, disturbed during the freezing process, and
from 0% to 3.3% in the largest studies retraction of the probes should not be
(71,74,77,78); however, incidence as high as attempted until the iceball has thawed, to allow
11 % (2/18) has been reported (80). probe removal with a minimum of force or
Other reported major complications follow- stress on the tissue.
ing prostate cryoablation include prostatic Other major, intraprocedural complications
abscess, testicular abscess (75), perirectal that have occurred include a tear in the splenic
abscess (80), sepsis, urethritis, epididymitis, capsule with the loss of approximately 1000mL
urinary tract infection, bladder outlet obstruc- of blood (83), and cryoinjury to the pancreas,
tion, bladder neck contracture, ureteral with subsequent pancreatitis from the inadver-
obstruction, urethral stricture, stone formation tent extension of the probe through and
in the prostatic urethra (81), prostatocutaneous beyond a lesion in the left kidney (88). Major,
fistula, rectovesical fistula (56), and vesi- postprocedural complications include chronic
courethral fistula (78). renal failure that necessitated hemodialysis
(83), superficial wound abscess (89), congestive
heart failure (83), dyspnea requiring 24-hour
Minor Complications
intubation, and paralytic ileus (87).
The most common minor complications of There has been concern that cryoablation of
prostate cryoablation include transient hema- lesions near the collecting system may result in
turia, rectal pain, pelvic pain, osteitis pubis (82), caliceal injury, leading to urinary extravasation
scrotal swelling, and penile tingling or numb- and possible fistula formation. However, there
ness. The rectal and pelvic pain frequently can have yet to be any reports of urine leakage or
be managed with nonsteroidal antiinflamma- fistula formation, despite the probable freezing
450 L. Cheung et al

of the collecting system in some patients. perforation with subsequent peritonitis and
Recent animal studies have demonstrated that adult respiratory distress syndrome (92). It is
the renal pelvis can be frozen with minimal risk unclear whether the perforation was due to
of injury (90,91). thermal damage suffered during the procedure
or from stress ulceration. Other cases of intesti-
Minor Complications nal perforation have not been reported. One
case of multisystem organ failure (95), and one
Reported minor complications following
death from presumed septic shock have been
renal cryoablation include small perirenal
reported (92). One patient with a known
hematomas (89), elevations of amylase and
history of cirrhosis and severe esophageal
lipase for 24 to 48 hours, asymptomatic elec-
varices died 2 weeks after UTI from massive
trocardiogram changes, transient atrial fibrilla-
variceal bleeding of the esophagus (92).
tion (88), small pelvic vein thrombosis, and
transient worsening of hemiparesis in a patient
Major Complications
with a prior left cerebrovascular accident (85).
The most common major complications follow-
ing hepatic UTI are pleural effusions that
Laser-Induced Interstitial require thoracentesis and hepatic abscess for-
Thermotherapy (LITT) mation. Pleural effusions are a common occur-
rence postprocedurally, and although most are
Hepatic small, asymptomatic, and resolve sponta-
neously, a small number that persist or cause
Most of the available data concerning compli- dyspnea require drainage. The risk of hepatic
cations following hepatic UTI are from studies abscess is significantly increased in patients
using a percutaneous approach for laser fiber who have undergone prior hepatobiliary
placement. There is less experience with UTT surgery, especially Whipple procedures for pan-
than with RFA or cryoablation; however, in the creatic adenocarcinoma (92).
few large studies that have been reported, mor- Other major complications that have been
tality from UTI is low, ranging from 0.3% to reported include injury to the major bile ducts,
1.0% (92,93). Major complications have segmental infarction from either coagulation of
occurred at rates of 3.2% to 4.7%, when based a major arterial vessel or from extensive laser-
on the number of patients treated (4,92); induced necrosis, intraperitoneal hemorrhage
however, many patients underwent multiple (92), systemic febrile reaction (96), paralytic
treatment sessions that indicate that the risk ileus, and acute liver failure in patients with
per UTT procedure is actually lower. Also, advanced cirrhosis (4). Intraprocedural brady-
LITI can be performed as an inpatient or out- cardia (heart rate <30) precluded the comple-
patient procedure, and one study comparing tion of one procedure (96). Only two cases of
complications in the two groups found that tumor seeding from the needle tract have been
there was no significant difference in the rates reported (94). In a study of 899 patients, there
of the two most common complications: pleural were no skin burns observed, which can be
effusion and subcapsular hematoma (92). partly attributed to the use of light protection
systems (LPS; Dornier Medizintechnik, Ger-
Mortality mering, Germany) with a fast automatic switch-
off function to prevent the fiberoptic bundles
Reported deaths following hepatic UTI
from being burned (92).
include two patients who developed acute liver
failure following the procedure, one from a
Minor Complications
massive hepatic infarction (94) and another
with Child-Pugh class C cirrhosis who died 2 The most common minor complications of
months after treatment (93). Another death hepatic UTI are pain, fever, nausea, and
occurred 4 weeks after treatment from jejunal asyptomatic pleural effusion. Other reported
36. Complications of Tumor Ablation 451

minor complications include abdominal wall surface of the liver and two on the abdominal
bruising, intrahepatic hematoma, subcapsular wall (99). Another case report described an
hematoma, subcutaneous hematoma, mild instance of generalized intraperitoneal seeding
intraperitoneal bleeding, transient biloma, local following hepatic MCT, with numerous small
wound infection, and pneumothorax requiring nodules found in the omentum, visceral serosa,
only intraprocedural evacuation (92-96). and in the pouch of Douglas (103).

Minor Complications
Microwave Coagulation The most commonly reported minor complica-
Therapy (MCT) tions following hepatic MCT are periproce-
dural pain and low-grade fever, which occur in
Hepatic a majority of patients. Other minor complica-
tions include pleural effusion, small subcapsu-
Data on the complications of hepatic MCT lar hematoma, subcutaneous hematoma,
are remarkably scarce. In one of the largest and minor skin burns. Clinically insignificant
reported series involving 234 patients, there hepatic infarction, pneumothorax (101), portal
were no deaths or major complications (97). vein thrombosis, and arterioportal shunt also
Overall, the rate of major complications ranges have been described (99).
from 0% to 11 % (97-101). In one study that
compared percutaneous and intraoperative
MCT, complications including hepatic abscess Conclusion
formation were much more common in the
intraoperative group (99). This could be due The popularity of tumor ablation has blos-
partly to a larger mean lesion size in patients somed, due, in part, to its favorable safety
for whom an open or laparoscopic approach profile compared to other treatment options.
was chosen (4.5cm) versus the percutaneous Although serious complications have occurred
approach (3.4cm). with ablation procedures, the risk of such
A prospective, randomized trial that com- events is acceptably low. Furthermore, the field
pared MCT and RFA for the treatment of HCC of tumor ablation is still relatively new, and
found that major complications occurred in 4 of there is a learning curve associated with it, as
36 patients (11 %) treated with MCT versus 1 with all procedures. Indeed, several long-term
of 36 (2.8%) for RFA (102). Although the mean studies have indicated that many more
lesion size was found to be similar between the complications occurred earlier in the respective
two groups, the number of treatment sessions authors' experience (17,92). It is expected that
per nodule was significantly higher in the MCT as proficiency in tumor ablation increases,
group than in the RFA group (2.4 vs. 1.1). This potential complications can be anticipated and
led to a prolonged treatment course in those averted, furthering the safety of ablation and
undergoing MCT. entrenching its role in therapeutic oncology.

Major Complications
References
The reported major complications following
hepatic MCT include acute liver failure (100), 1. Bret PM, Sente JM, Bretagnolle M, et al. Ultra-
sonically guided fine-needle biopsy in focal
ascites, intraperitoneal hemorrhage (101), sub-
intrahepatic lesions: 6 years experience. Can
capsular hematoma, liver abscess, subcutaneous
Assoc Radiol J 1986;37:5-8.
abscess (102), cholangitis, persistent fever 2. Di Stasi M, Buscarini L, Livraghi T, et al. Per-
(100), and needle-tract seeding. Although the cutaneous ethanol injection in the treatment of
incidence of needle-tract seeding generally is hepatocellular carcinoma. A multicenter survey
low following MCT, one study found three of evaluation practices and complication rates.
cases out of 63 patients (4.8%)-one on the Scand J GastroenteroI1997;32:1168-1173.
452 L. Cheung et al

3. Livraghi T, Giorgio A, Marin G, et al. Hepato- 16. Angelini F, Nacamulli D, De Vido D, et al. Treat-
cellular carcinoma and cirrhosis in 746 patients: ment of hot thyroid nodule with percutaneous
long-term results of percutaneous ethanol ethanol injection: indications, complications,
injection. Radiology 1995;197:101-108. and prognostic factors. Radiologia Medica 1996;
4. Giorgio A, Tarantino L, de Stefano G, et al. 91:774-780.
Complications after interventional sonography 17. Livraghi T, Solbiati L, Meloni MF, et al. Treat-
of focal liver lesions. A 22-year single-center ment of focal liver tumors with percutaneous
experience. J Ultrasound Med 2003;22:193-205. radio-frequency ablation: complications encoun-
5. Livraghi T, Vettori C, Torzilli G, et al. Long-term tered in a multicenter study. Radiology 2003;
results of single-session percutaneous ethanol 226:441-451.
injection in patients with large hepatocellular 18. de Baere T, Risse 0, Kuoch V, et al. Adverse
carcinoma. Cancer 1998;83:48-57. events during radiofrequency treatment of 582
6. Boucher E, Carsin A, Raoul JL, et al. Massive hepatic tumors. AJR. Am J Roentgenol 2003;
hepatic necrosis secondary to treatment of 181:695-700.
hepatocellular carcinoma by percutaneous 19. Schmidt-Mutter C, Breining T, Gangi A, et al.
alcoholization. Gastroenterol Clin BioI 199;22: Fatal bile pulmonary embolism after radiofre-
459-461. quency treatment of a hepatocellular carci-
7. de Baere T, Roche A,Amenabar JM, et al. Liver noma. Surg Endosc 2003;17:2028-2031.
abscess formation after local treatment of liver 20. de Sio I, Castellano L, De Girolamo V, et al.
tumors. Hepatology 1996;23:1436-1440. Thmor dissemination after radiofrequency
8. Miki K, Makuuchi M, Takayama T, et al. Peri- ablation of hepatocellular carcinoma. Hepatol-
toneal seeding of hepatocellular carcinoma ogy 2001;34:609-610.
after ethanol injection therapy. Hepato- 21. Llovet JM, Vilana R, Bm C, et al. Increased risk
Gastroenterology 2000;47:1428-1430. of tumor seeding after percutaneous radiofre-
9. Ishii H, Okada S, Okusaka T, et al. Needle tract quency ablation for single hepatocellular carci-
implantation of hepatocellular carcinoma after noma. Hepatology 2001;33:1124-1129.
percutaneous ethanol injection. Cancer 1998; 22. Chapman W, Debelak J, Pinson CW, et al.
82:1638-1642. Hepatic cryoablation, but not radiofrequency
10. Casella G, Cacopardo E, Rovere G, et al. Cuta- ablation, results in lung inflammation. Ann Surg
neous seeding after ultrasound-guided per- 2000;231:752-761.
cutaneous ethanol injection for treatment of 23. Seifert J, France M, Zhao J, et al. Large volume
hepatocellular carcinoma. J Clin Ultrasound hepatic freezing: association with significant
2001 ;29:354-358. release of the cytokines interleukin-6 and
11. Matsumata T, Okamoto M, Ishikawa H, et al. tumor necrosis factor-alpha in a rat model.
Necrosis of the hepatocellular carcinoma World J Surg 2002;26:1333-1341.
nodule can aggravate metastasis. Nippon Geka 24. Yamamoto T, Kubo S, Hirohashi K, et al.
Gakkai Zasshi 1998;99:197-200. Secondary hemocholecyst after radiofrequency
12. Elgindy N, Lindolm H, Gunven P. High-dose ablation therapy for hepatocellular carcinoma.
percutaneous ethanol injection therapy of liver J Gastroenterol 2003;28:399-403.
tumors. Patient acceptance and complications. 25. Verhoeven B, Haagsma E, Appeltans B, et al.
Acta Radiologica 2000;41:458-463. Hyperkalaemia after radiofrequency ablation
13. Seki S, Kitada T, Otogawa K, et al. Gastric ulcer of hepatocellular carcinoma. Eur J Gastroen-
as a rare complication of percutaneous ethanol terol HepatoI2002;14:1023-1024.
injection for hepatocellular carcinoma. Am J 26. Onik G, Onik C, Medary I, et al. Life-
GasteroenteroI2002;97:77D-771. threatening hypertensive crises in two patients
14. Isobe H, Sakamoto S, Nawata H. Splenic undergoing hepatic radiofrequency ablation.
abscess as a complication of percutaneous AJR Am J RoentegenoI2003;181:495-497.
ethanol injection therapy for hepatocellular 27. Napier D, Dodd G, Hubbard L, et al. Post-
carcinoma. J Clin Gastroenterol 1992;15:167- ablation syndrome following radiofrequency
179. thermal ablation of liver tumors. Presented at
15. Komorowski J, Kuzdak K, Pomorski L, et al. SGR 29th Annual Meeting. March 12, 2000.
Percutaneous ethanol injection in treatment Kauai, Hawaii.
of benign nonfunctional and hyperfunctional 28. Goldberg SN, S9.lb~ati L, Halpern EF, et al. Vari-
thyroid nodules. Cytobios 1998;95:143-150. ables affecting I p~oper system grounding for
36. Complications of Tumor Ablation 453

radiofrequency ablation in an animal model. ablation of solid renal masses: techniques and
J Vasc Interv Radio12000;11:1069-1075. outcomes of 38 treatment sessions in 32 con-
29. King J, Glenn D, Clark W, et al. Percutaneous secutive patients. AJR Am J Roentgenol 2003;
radiofrequency ablation of pulmonary metas- 180:1503-1508.
tases in patients with colorectal cancer. Br J 41. Johnson D, Saboorian M, Duchene D, et a1.
Surg 2004;91:217-223. Nephrectomy after radiofrequency ablation-
30. vanSonnenberg E, Shanker S, Morrison PR, et induced ureteropelvic junction obstruction:
al. Radiofrequency ablation of thoracic lesions: potential complication and long-term assess-
part II initial clinical experience-technical and ment of ablation adequacy. Urology 2003;62:
multidisciplinary considerations in 30 patients. 351-352.
AJR 2005;184:381-390. 42. Goetz M, Callstrom M, Charboneau J, et al.
31. Dupuy D, Mayo-Smith W, DiPetrillo T, et a1. Percutaneous image-guided radiofrequency
Clinical experience of pulmonary radiofre- ablation of painful metastases involving bone:
quency ablation in 27 patients. Radiological a multicenter study. J Clin Oncol 2004;22:
Society of North America Proceedings, 300-306.
Chicago, November 2001;Abstract 389. 43. Ibrahim D, Ho E, Scher! SA, et a!. Newborn
32. Nishida T, Inoue K, Kawata Y, et a1. Per- with an open posterior hip dislocation and
cutaneous radiofrequency ablation of lung sciatic nerve injury after intrauterine radiofre-
neoplasms: a minimally invasive strategy for quency ablation of a sacrococcygeal teratoma.
inoperable patients. J Am ColI Surg 2002;195: J Pediatr Surg 2003;38:248-250.
426-430. 44. Wood B, Abraham J, Hvizda J, et a1. Radiofre-
33. Sewell P Jr, Jackson M, Vance R, et a1. quency ablation of adrenal tumors and adreno-
Assessing radiofrequency ablation of non-small cortical carcinoma metastases. Cancer 2003;97:
cell lung cancer with positron emission 554-560.
tomography (PET). Radiology 2000;217 45. Mayo-Smith W, Dupuy D. Adrenal neoplasms:
(Suppl):334. CT-guided radiofrequency ablation-prelimi-
34. Lee J, Jin K, Kim C, et al. Percutaneous nary results. Radiology 2004;231:225-230.
radiofrequency ablation for inoperable lung 46. Izzo F, Thomas R, Delrio P, et al. Radiofre-
malignancies: a preliminary report. Radiologi- quency ablation in patients with primary breast
cal Society of North America Proceedings, carcinoma: a pilot study in 26 patients. Cancer
Chicago, November 2001;Abstract 390. 2001;92:2036-2044.
35. Vaughn C, Mychaskiw G II, Sewell P. Massive 47. Burak W,Agnese D, Povoski S, et al. Radiofre-
hemorrhage during radiofrequency ablation of quency ablation of invasive breast carcinoma
a pulmonary neoplasm. Anesth Analg 2002;94: followed by delayed surgical excision. Cancer
1149-1151. 2003;98:1369-1375.
36. Gervais D, McGovern F, Wood B, et al. Radio- 48. Hayashi A, Silver S, van der Westhuizen N,
frequency ablation of renal cell carcinoma: et a1. Treatment of invasive breast carcinoma
early clinical experience. Radiology 2000;217: with ultrasound-guided radiofrequency abla-
665-672. tion. Am J Surg 2003;185:429-435.
37. Pavlovich C, McClellan W, Choyke P, et a1. Per- 49. Fornage B, Sneige N, Ross M, et a1. Small (:::2-
cutaneous radio frequency ablation of small cm) breast cancer treated with US-guided
renal tumors: initial results. J Urol 2002;167: radiofrequency ablation: feasibility study. Radi-
10-15. ology 2004;231:215-224.
38. de Baere T, Kuoch V, Smayra T, et a1. Radio fre- 50. Adam R, Hagopian E, Linhares M, et al. A
quency ablation of renal cell carcinoma: pre- comparison of percutaneous cryosurgery and
liminary clinical experience. J Urol 2002;167: percutaneous radiofrequency for unresectable
1961-1964. hepatic malignancies. Arch Surg 2002;137:
39. Su L, Jarrett T, Chan D, et al. Percutaneous 1332-1339.
computed tomography-guided radiofrequency 51. Tait LS, Yong SM, Cuschieri A. Laparoscopic in
ablation of renal masses in high surgical risk situ ablation of liver cancer with cryotherapy
patients: preliminary results. Urology 2003; and radiofrequency ablation. Br J Surg 2002;
61(Supp14A):26-33. 89:1613-1619.
40. Mayo-Smith W, Dupuy D, Parikh P, et al. 52. Lezoche E, Paganini A, Feliciotti F, et a1.
Imaging-guided percutaneous radiofrequency Ultrasound-guided laparoscopic cryoablation
454 L. Cheung et al

of hepatic tumors: preliminary report. World J 68. Onik G, Cobb C, Cohen J, et al. US character-
Surg 1998;22:829-836. istics of frozen prostate. Radiology 1988;168:
53. Sarantou T, Bilchik A, Ramming K. Complica- 629-631.
tions of hepatic cryosurgery. Semin Surg Oncol 69. Cohen J, Miller R. Thermal protection of
1998;14:156-162. urethra during cryosurgery of prostate. Cryobi-
54. Sohn R, Carlin A, Steffes C, et al. The extent ology 1994;31:313-316.
of cryosurgery increases the complication rate 70. Zisman A, Pantuck A, Cohen 1. Prostate
after hepatic cryoablation. Am Surg 2003;69: cryoablation using direct transperineal place-
317-322. ment of ultrathin probes through a 17-gauge
55. Crews K, Kuhn J, McCarty T, et al. Cryosurgi- brachytherapy template: technique and prelim-
cal ablation of hepatic tumors. Am J Surg inary results. Urology 2001;58:988-993.
1997;174:614-617. 71. Han K, Cohen J, Miller R, et al. Treatment of
56. Seifert J, Morris D. World survey on the com- organ-confined prostate cancer with 3rd gen-
plications of hepatic and prostate cryotherapy. eration cryosurgery: preliminary multi-center
World J Surg 1999;23:109-114. experience. J Urol2003;170:1126-1130.
57. Cha C, Lee F, Rikkers L, et al. Rationale for 72. Long J, Bahn D, Lee F, et al. Five-year retro-
the combination of cryoablation with surgical spective, multi-institutional pooled analysis of
resection of hepatic tumors. J Gastrointest Surg cancer-related outcomes after cryosurgical
2001 ;5:206-213. ablation of the prostate. Urology 2001;57:518-
58. Onik G, Atkinson D, Zemel R, et al. 523.
Cryosurgery of liver cancer. Semin Surg Oncol 73. Bahn D, Lee F, Badalament R, et al. Targeted
1993;9:309-317. cryoablation of the prostate: 7-year outcomes
59. Pearson AS, Izzo P, Fleming RY, et al. Intra- in the primary treatment of prostate cancer.
operative radiofrequency ablation or cryoabla- Urology 2002;60(2 Suppl1):3-11.
tion for hepatic malignancies. Am J Surg 1999; 74. Pisters L, von Eschenbach A, Scott S, et al. The
178:592-599. efficacy and complications of salvage cryother-
60. Morris D, Ross W, Iqbal J, et al. Cryoablation apy of the prostate. J Urol1997;157:921-925.
of hepatic malignancy: an evaluation of tumor 75. Donnelly B, Saliken J, Ernst D, et al. Pros-
marker data and survival in 110 patients. Gas- pective trial of cryosurgical ablation of the
trointest Cancer 1996;1:247-251. prostate: five-year results. Urology 2002;60:
61. Seifert J, Stewart G, Hewitt P, et al. Interleukin- 645-649.
6 and tumor necrosis factor-alpha levels fol- 76. Onik G, Narayan P, Vaughan D, et al. Focal
lowing hepatic cryotherapy: association with "nerve-sparing" cryosurgery for treatment of
volume and duration of freezing. World J Surg primary prostate cancer: a new approach
1999;23:1019-1026. to preserving potency. Urology 2002;60:109-
62. Yeh K, Fortunato L, Hoffman J, et al. Cryosur- 114.
gical ablation of hepatic metastases from colo- 77. Ghafar M, Johnson C, De La Taille A, et al.
rectal carcinomas. Am Surg 1997;63:63-68. Salvage cryotherapy using an argon based
63. Weaver ML, Ashton J, Zemel R. Treatment of system for locally recurrent prostate cancer
colorectal liver metastases by cryotherapy. after radiation therapy: the Columbia experi-
Semin Surg Oncol1998;14:163-170. ence. J UroI2001;166:1333-1338.
64. Silverstein JC, Staren E, Velasco 1. Thermal bile 78. Chin J, Pautler S, Mouraviev V, et al. Results
duct protection during liver cryoablation. J Surg of salvage cryoablation of the prostate after
OncoI1997;64:163-164. radiation: identifying predictors of treatment
65. Weaver ML, Atkinson D, Zemel R. Hepatic failure and complications. J Urol 2001;165:
cryosurgery in treating colorectal metastases. 1937-1942.
Cancer 1995;76:210--214. 79. Wieder J, Schmidt JD, Casola G, et al. Trans-
66. Cozzi P, Stewart G, Morris D. Thrombocytope- rectal ultrasound-guided transperineal cryoab-
nia after hepatic cryotherapy for colorectal lation in the treatment of prostate carcinoma:
metastases: correlates with hepatocellular preliminary results. J UroI1995;154:435-441.
injury. World J Surg 1994;18:774-777. 80. Long J, Fallick M, LaRock D, et al. Preliminary
67. Gonder M, Soanes W, Shulman S. Cryosurgical outcomes following cryosurgical ablation of
treatment of the prostate. Invest Urol 1966;3: the prostate in patients with clinically localized
372-378. prostate carcinoma. J UroI1998;159:477-484.
36. Complications of Tumor Ablation 455

81. Aus G, Bergdahl S, Hugosson J, et al. Stone 94. Gilliams A, Lees W. Survival after percuta-
formation in the prostatic urethra after neous, image-guided, thermal ablation of
cryotherapy for prostate cancer. Urology 1997; hepatic metastases from colorectal cancer. Dis
50:615-617. Colon Rectum 2000;43:656-661.
82. Seigne J, Pisters L, von Eschenbach AC. Osteitis 95. Tranberg K, Moller P, Hannesson P, et al. Inter-
pubis as a complication of prostate cryotherapy. stitial laser treatment of malignant tumors:
J Urol 1996;156:182. initial experience. Eur J Surg Oncol 1996;22:
83. Rukstalis D, Khorsandi M, Garcia F, et al. Clin- 47-54.
ical experience with open renal cryoablation. 96. Christophi C, Muralidharan V. Chronic liver
Urology 2001;57:34-39. diseases: epidemiology, pathophysiology, diag-
84. Delworth M, Pisters L, Fornage B, et al. nosis and treatment. J Gastroenterol Hepatol
Cryotherapy for renal cell carcinoma and 2001;16:548-552.
angiomyolipoma. J Urol 1996;155:252-255. 97. Dong B, Liang P, Yu X, et al. Percutaneous
85. Rodriguez R, Chan D, Bishoff J, et al. Renal sonographically guided microwave coagulation
ablative cryosurgery in selected patients with therapy for hepatocellular carcinoma: results in
peripheral renal masses. Urology 2000;55:25-30. 234 patients. AJR Am J Roentgenol 2003;180:
86. Gill I, Novick A, Meraney A, et al. Laparo- 1547-1555.
scopic renal cryoablation in 32 patients. 98. Liang P, Dong B, Yu X, et al. Prognostic factors
Urology 2000;56:748-753. for percutaneous microwave coagulation
87. Nadler R, Kim S, Rubenstein J, et al. Laparo- therapy of hepatic metastases. AJR Am J
scopic renal cryosurgery: the Northwestern RoentgenoI2003;181:1319-1325.
experience. J UroI2003;170:1121-1125. 99. Mitsuzaki K, Yamashita Y, Nishiharu T, et al. CT
88. Lee D, McGinnis D, Feld R, et al. Retroperi- appearance of hepatic tumors after microwave
toneallaparoscopic cryoablation of small renal coagulation therapy. AJR Am J Roentgenol
tumors: intermediate results. Urology 2003;61: 1998;171:1397-1403.
83-88. 100. Yamanaka N, Tanaka T, Oriyama T, et al.
89. Shingleton W, Sewell P. Percutaneous renal Microwave coagulonecrotic therapy for hepa-
tumor cryoablation with magnetic resonance tocellular carcinoma. World J Surg 1996;20:
image guidance. J UroI2001;165:773-776. 1076-1081.
90. Chosy S, Nakada S, Lee F, et al. Monitoring 101. Abe T, Shinzawa H, Wakabayashi H, et al. Value
renal cryosurgery: predictors of tissue necrosis of laparoscopic microwave coagulation therapy
in swine. J UroI1998;159:1370-1374. for hepatocellular carcinoma in relation to
91. Sung G, Gill I, Hsu T, et al. Effect of intentional tumor size and location. Endoscopy 2000;32:
cryoinjury to the renal collecting system. 598--603.
J EndouroI1999;13(S1):A14. 102. Shibata T, Iimuro Y, Yamamoto Y, et al. Small
92. Vogi T, Straub R, Eichler K, et al. Malignant hepatocellular carcinoma: comparison of radio-
liver tumor treated with MR imaging-guided frequency ablation and percutaneous micro-
laser-induced thermotherapy: experience with wave coagulation therapy. Radiology 2002;223:
complications in 899 patients (2520 lesions). 331-337.
Radiology 2002;225:367-377. 103. Sato M,Tokui K, Watanabe Y, et al. Generalized
93. Giorgio A, Tarantino L, de Stefano G, et al. intraperitoneal seeding of hepatocellular carci-
Interstitial laser photocoagulation under ultra- noma after microwave coagulation therapy: a
sound guidance of liver tumors: results in 104 case report. Hepato-Gastroenterol 1999;46:
treated patients. Eur J Ultrasound 2000;11:181- 2561-2564.
188.
Section VI
Perspectives
37
Tumor Ablation for Patients with Lung
Cancer: The Thoracic Oncologist's
Perspective
Bruce E. Johnson and Pasi A. Janne

Lung cancer is the second most frequent cause there is no lymph node or metastatic cancer
of cancer in both men and women in the United involvement (2,3). Some patients have inade-
States (1). There were an estimated 91,800 lung quate respiratory reserve to tolerate surgical
cancer cases in men and 80,100 cases in women resection of their lung cancer. There is an exten-
in the United States in 2003. The most effec- sive experience in treating these patients with
tive treatment for patients with localized lung radiotherapy to the primary lesion in the lung;
cancer is surgical resection. Radiation therapy approximately 20% of these patients are alive
and chemotherapy also are established effec- 5 years after the start of the radiotherapy (4,5).
tive treatments in patients with more advanced However, only approximately 26% of patients
lung cancer. The role of tumor ablation in the presenting with lung cancer have their cancer
management of patients with lung cancer is localized to single lobe and without any evi-
evolving. To understand the current and future dence of spread to the lymph nodes in the
potential role in the management of patients mediastinum or bloodstream. Most patients are
with lung cancer, it is important to have infor- unable to be treated with these effective local
mation about the available therapies for the dif- therapies alone (6).
ferent stages of lung cancer. Patients with lung cancer most commonly
present when the cancer has spread locally in
the chest, into the lymph nodes in the
mediastinum (stage III), and systemically
Conventional Treatment for through the lymphatic system or bloodstream
Patients with Lung Cancer (stage IV). Patients with cancer spread to the
structures in the chest or lymph nodes in the
Treatment of lung cancer is effective if the lung mediastinum are most commonly treated with
cancer is detected when it has not spread locally mUltiple therapeutic modalities. These repre-
to the structures in the chest wall, mediastinum, sent approximately 33% of patients who
bones, or the lymph nodes in the mediastinum develop lung cancer. Fit patients with stage III
(stages I and II). Patients should be treated lung cancer are treated with combination
with surgical resection by removing the chemotherapy and chest irradiation (7-9). The
affected lobe or lung if they can physiologically therapy is much less effective than surgical
tolerate the removal of one to three of the five treatment of patients with stage I and II
lobes of their lungs. Seventy percent to 80% of non-small-cell lung cancer because patients
these patients with the earliest stages of lung with stage III lung cancer have an expected 5-
cancer are alive and free of lung cancer at 5 year survival of 5% to 15%. The therapies cur-
years if the lung cancer is less than 3 cm and rently available to patients with lung cancer

459
460 B.E. Johnson and P.A. Janne

that has spread through the bloodstream are Animal Model of Lung Cancer
relatively ineffective. Approximately 41 % of
patients present with lung cancer that has
Treated with Radiofrequency
spread through the lymphatic system or blood- Ablation
stream. The use of combination chemotherapy
for patients with stage IV lung cancer evokes An animal model has been developed to test the
response rates in 20%, the median survival is safety and efficacy of RFA in rabbits. The VX2
7 to 9 months (1 to 2 months longer than in carcinoma cell line is injected into the lungs of
patients who are not treated with combination New Zealand rabbits. The growth of the lung
chemotherapy), and only 2% of patients are tumor(s) can be monitored by computed
alive 5 years after the start of their chemother- tomography or magnetic resonance imaging.
apy (10-13). The lung tumors reach a size of 1 cm in a few
weeks (19-21). The cancers then grow within the
lungs and spread systemically. The rabbits go on
Development of Radiofrequency to die from their cancer within 3 months in the
absence of any treatment. The course of these
Ablation for Patients with cancers growing within the lung can be altered
Lung Cancer by treatment with RFA. Twenty-five rabbits
have been treated with a radiofrequency gener-
There is more than 10 years of experience ator capable of producing 150W of power
using radiofrequency to ablate metastatic and (Radionics, Burlington, MA). The probe of the
primary cancers in the liver; extensive follow- generator was passed percutaneously through
up of these patients after this treatment has the chest wall and into the lesion. The size of the
documented its efficacy (14-18). Research in lesions and position of the probe were verified
this area has led to the development of probes by imaging. The tumor was ablated by applying
that can ablate tumors greater than 1 cm, which power to the radiofrequency generator for
is important for the treatment of lung cancers several minutes as determined by size of the
(18). lesions and previous experimental results. The
The success of this percutaneous approach treatment caused complications in the rabbits:
has prompted developing techniques to use two rabbits developed significant pneumotho-
radiofrequency ablation (RFA) in the lung for races requiring surgery or euthanasia, and a
both primary lung lesions and metastases. third rabbit developed a pyothorax after being
There are currently several potential uses for treated with RFA.
tumor ablation in patients with lung cancer Twenty-three of the rabbits were sacrificed
including those with lung cancer whose lung a few hours to 2 months after treatment with
function is not adequate to tolerate a surgical RFA. Histologic examination of the tumors in
resection or localized radiation therapy. The the lungs showed necrosis in the lesions within
other common indications are inpatients who a few hours and eventual eradication of the
have a local problem either in the initial cancer tumor in most rabbits after the treatment.
or in a metastatic site that cannot be irradiated One of the experiments followed the animals
or undergo surgical resection. This can be due for several months after their treatment (19).
either to the location of the tumor, or to the Rabbits were sacrificed and their tumors exam-
growth of the tumor after primary radiation ined histologically. More than half of the rabbits
therapy. The local problems can include devel- had their tumors eradicated. Four rabbits
opment of a metachronous lung cancer, lobar treated with RFA were not sacrificed and lived
obstruction, pain caused by localized tumor longer than 3 months, the time when all rabbits
masses, obstruction caused by a tumor mass, or inoculated with VX2 and not treated with RFA
isolated metastatic disease. had died from their cancer. The information
37. Thmor Ablation for Patients with Lung Cancer 461

generated from this animal model and infor- will be alive 5 or more years after this treatment
mation from treating patients with primary (4,5).
liver cancer and liver metastases supported For those patients who are unable or un-
going forward into treatments of lung tumors in willing to undergo surgery and/or radiation
humans with RFA. therapy, local treatment including RFA may be
considered. Intraparenchymal cancers in the
lung may well be suited to RFA because the air
in the surrounding pulmonary parenchyma
Tumor Ablation of Primary and provides insulation around the cancer (23).
Metachronous Lung Cancers Therefore, the energy generated from the
radiofrequency probe may be concentrated in
The standard treatment of lung cancer localized the lesions, while having relatively little effect
to the lung without evidence of spread to the in the surrounding tissues. However, some
lymph nodes or through the bloodstream is sur- tumor masses near major blood vessels or the
gical resection. However, not all patients can heart are not appropriate candidates for tumor
tolerate a pulmonary resection. Approximately ablation because of potential damage to the
90% of patients with lung cancer are current or vascular structures that might lead to bleeding
former cigarette smokers with some degree of complications. Figure 37.1 shows a lesion invad-
pulmonary impairment. In addition, patients ing blood vessels and Figure 37.2 shows a lesion
developing lung cancer in the United States near the heart and aorta.
usually are elderly with a median age of 69 There are examples in the literature of pa-
years and comorbid diseases that preclude tients whose primary lung cancers, synchronous
surgical resection. Patients with severe pul- lung cancers, or metachronous lung cancers
monary impairment or comorbid diseases have been treated with RFA and have not
should undergo a careful evaluation by an
experienced general thoracic surgeon to deter-
mine if they should have an attempt at surgical
resection.
The other group of patients who are poten-
tial candidates are those who had undergone
a successful surgical resection of their non-
small-cell lung cancer. These patients may have
one to three lobes removed to resect their
non-small-celliung cancer adequately. Patients
treated successfully for non-small-cell lung
cancer have a 2% risk per year of developing a
second lung cancer in their remaining lung (22).
These patients have diminished pulmonary
reserve and may not be able to undergo further
resections. Patients who are unable to undergo
an initial or repeat surgical resection should
be referred to radiation oncologists. There is
substantial literature about patients who were FIGURE 37.1. Non-small-cell lung carcinoma with
vascular invasion. The patient presented with a
unable or unwilling to undergo a surgical
tumor in the left lung that could not be resected due
resection and who then are treated with radia-
to poor pulmonary reserve. The patient was treated
tion therapy to the primary lesions. Doses of with primary radiation therapy and 12 months later
approximately 6000 cGy are administered to the tumor began to enlarge. Thmor ablation was con-
the primary tumor mass. Approximately 20% of sidered, but not attempted due to vascular invasion
patients with stage I and II non-small-celliung (arrows) of the tumor mass near large branches of
cancer treated with chest radiotherapy alone the pulmonary artery.
462 RE. Johnson and P.A. Hnne

recurred after follow-up of 1 to 3 years (Table


37.1). The information thus far is anecdotal,
with approximately 10 patients reported in the
peer-reviewed published literature. Thus, it is
yet another local therapy that can be employed
if surgical resection and/or radiation therapy
are deemed to be inappropriate and the cancer
can be controlled for an extended period of
time. Further research will be needed to define
the role of tumor ablation in these patient
populations.
There are potential complications associated
with the RFA. Dupuy et al (23) reported pneu-
mothoraces in 20% of patients treated with
RFA with most requiring chest tube placement.
This is an important potential complication,
because patients who undergo RFA because
FIGURE 37.2. Metastatic mesothelioma adjacent to
the heart. The patient was treated with a right-sided
of poor pulmonary reserve and then develop
extrapleural pneumonectomy, but developed con- significant pneumonthorax may develop life-
tralateral pulmonary metastases. Thmor ablation of threatening pulmonary insufficiency. Therefore,
the mass adjacent to the heart (arrow) was consid- the procedure should be performed in a setting
ered, but deferred because of the proximity to the in which patients can be treated with res-
heart. piratory support and emergent chest tube
placement. Other side effects include pleurisy,
development of a pleural effusion, and a pro-

TABLE 37.1. Literature review on patients with lung cancer treated with radiofrequency ablation.
Patient Previous
(reference) Age Stage treatment Reason for ablation Outcome
1. Male (23) 78 III None Shrink mass before Alive at 27 months
chest RT
2. Male (23) 75 Metachronous stage I Pneumonectomy Eradicate nodule Alive 11 months
plus brachytherapy
3. Female (23) 69 I None Eradicate nodule Alive at 24 months
4. Male (23) I None Eradicate nodule Alive 1 week later
5. Male (24) 70 IV Surgery and Eradicate nodule Died from complications
chemotherapy of bleeding within
1 month
6. Male (28) 59 2 stage I NSCLC None Eradicate nodules Alive at 10 months
(metachronous in
RULand RLL)
7. Male (29) 45 IV Chemotherapy Eradicate nodule Alive at 3 months
and chest RT
8. Female (29) 66 IE None Shrink nodule Died at nursing home 2
before external RT months after procedure
9. Male (30) 56 Metachronous Surgery Eradicate nodule Alive at 2 years
lung cancers
10. Male (30) 72 Metachronous Surgery Eradicate nodule Alive at 1 year
lung cancer

Abbreviations: RT, chest radiotherapy; RLL, right lower lobe; RUL, right upper lobe; NSCLC, nonsmall cell lung cancer.
37. Thmor Ablation for Patients with Lung Cancer 463

ductive cough. Patients also can develop bleed- ment with RFA. Thus, this is one of many
ing following treatment with tumor ablation. A methods to relieve bronchial obstruction.
patient was treated with RFA of a lesion in the Further research will be needed to determine
right lung (patient number 5, Table 37.1). Two which of the many modalities of treatment is
hours after the procedure he developed pul- appropriate for different clinical situations.
monary hemorrhage and eventually died from Tumor ablation also has been used for
complications within the next month (24). The patients developing a local problem related to
patient was being treated with an antiplatelet tumor masses. This can be due to contiguous
agent (clopidogrel) that may have contributed spread to the bone from an underlying lung
to this bleeding complication. The authors rec- cancer that can give rise to severe bone pain.
ommend that patients be questioned closely One early report documents a patient with lung
about the use of antiplatelet agents before cancer that spread to the sternum, and the
attempting an ablative procedure. patient underwent external irradiation; the lung
cancer had grown in the previously irradiated
area and caused bone pain (23). He was treated
Tumor Ablation of Locally with RFA with subsequent relief of the pain.
Our multidisciplinary group also has utilized
Recurrent or Metastatic RFA in patients with metastatic lung cancer.
Lung Cancer One example is a patient who has been treated
for osseous pain from a lesion in the pulmonary
Patients with non-small-cell lung cancer parenchyma that eroded into both ribs and ver-
treated with radiation can redevelop sympto- tebra (Fig. 37.3). Another patient developed a
matic cancer progression in the same area. localized relapse in the left adrenal gland and
Patients with stage III non-small-cell lung liver 6 years after treatment of the primary lung
cancer will relapse within the radiation portal cancer (Fig. 37.4). The patient was treated with
approximately 20% of the time (25). This can RFA of the adrenal and liver lesion. These are
rarely be treated with surgical resection or a
second course of radiation therapy. These
patients are potential candidates for percuta-
neous or endobronchial RFA if there is ade-
quate distance from the vital structures of the
pulmonary artery, aorta, and heart to the lesion.
A common local problem caused by lung
cancer is bronchial obstruction. This can occur
in approximately 20% of patients with lung
cancer at the time of presentation (26) and a
greater percentage in patients followed during
their disease course. The bronchial obstruction
can be treated with external irradiation, endo-
bronchial irradiation, stenting, photodynamic
therapy, cryotherapy, laser therapy, or RFA.
Radiofrequency ablation has been used com-
monly to treat patients with bronchial obstruc-
tion. There has been a series of 94 patients with
lung cancer treated with RFA including 50 FIGURE 37.3. Patient developed a mass in the periph-
with a previous course of chest irradiation or ery of the lower lobe that caused back pain. The
chemotherapy (27). Forty-six of the 50 patients probe is shown in place in the middle of the mass.
were able to have more than 50% of the The patient had partial relief of the pain following
luminal obstruction cleared following treat- the procedure.
464 RE. Johnson and P.A. Janne

References

1. Jemal A, Murray T, Samuels A, Ghafoor A, Ward


E, Thun MJ. Cancer statistics, 2003. CA Cancer J
Clin 2003;53:5-26.
2. Mountain CF. Revisions in the international
system for staging lung cancer. Chest 1997;111:
1710-1717.
3. Naruke T, Tsuchiya R, Kondo H, Asamura H.
Prognosis and survival after resection for bron-
chogenic carcinoma based on the 1997 TNM-
staging classification: the Japanese experience.
Ann Thorac Surg 2001;71:1759-1764.
4. Gauden S, Ramsay J, Tripcony L. The curative
treatment by radiotherapy alone of stage I non-
FIGURE 37.4. Patient presented with enlarging right small cell carcinoma of the lung. Chest 1995;108:
adrenal and liver lesions 6 years after resection of 1278-1282.
her T3NOMO non-small-cell lung cancer. The lesions 5. Langendijk JA, Aaronson NK, de Jong JM, et al.
continued to grow during multiple courses of Quality of life after curative radiotherapy in
chemotherapy. Metastatic evaluation showed no stage I non-small-cell lung cancer. Int J Radiat
other evidence of cancer, so she underwent radio- Oncol BioI Phys 2002;53:847-853.
frequency ablation. This view shows planning of the 6. Fry WA, Phillips JL, Menck HR. Ten-year survey
probe into the adrenal and liver lesion. The treat- of lung cancer treatment and survival in hospi-
ment has controlled the lesion 3 months after the tals in the United States: a national cancer data
radiofrequency ablation. base report. Cancer 1999;86:1867-1876.
7. Dillman RO, Herndon J, Seagren SL, Eaton WL,
Green MR. Improved survival in stage III
non-small-cell lung cancer: seven-year follow-up
of Cancer and Leukemia Group B (CALGB)
anecdotal examples of effective RFA treatment 8433 Trial. J Nat Cancer Inst 1996;88:1210-1215.
of painful bony lesions and ablative treatment 8. Furuse K, Fukuoka M, Kawahara M, et al. Phase
of isolated recurrences. III study of concurrent versus sequential tho-
Thus, tumor ablation using radiofrequency racic radiotherapy in combination with mito-
has been applied by percutaneous application mycin, vindesine, and cisplatin in unresectable
stage III non-small-cell lung cancer. J Clin Oncol
as well as by endobronchial administration.
1999;17:2692-2699.
Both have been shown to be effective therapies 9. Sause WT, Scott C, Taylor S, et al. Radiation
for some localized pulmonary lesions, as well as Therapy Oncology Group (RTOG) 88-08 and
endobronchial tumors. Radiofrequency abla- Eastern Cooperative Group (ECOG) 4588: pre-
tion also can playa role in metastatic or recur- liminary results of a phase III trial in regionally
rent lesions in areas not amenable to surgery or advanced, unresectable non-small cell lung
radiation therapy (32,33). This can include cancer. J Natl Cancer Inst 1995;87:198-205.
relief of painful bony lesions or isolated metas- 10. Group N-SCLCC. Chemotherapy in non-small
tases to the liver. The role of this local modal- cell lung cancer: a meta-analysis using updated
ity in patients who are unable to undergo data on individual patients from 52 randomized
surgery and/or irradiation likely will expand. clinical trials. Br Med J 1995;311:899-909.
11. Kelly K, Crowley J, Bunn PA Jr, et al. Random-
Further clinical research will be needed to
ized phase III trial of paclitaxel plus carboplatin
define the role of RFA in patients with airway
versus vinorelbine plus cisplatin in the treatment
obstruction in relationship to cryotherapy, of patients with advanced non-small-cell lung
laser treatment, and photodynamic therapy. cancer: a Southwest Oncology Group trial. J Clin
Further clinical research also will be needed to OncoI2001;19:3210-3218.
define its role for management of metastatic 12. Schiller JH, Harrington D, Belani CP, et al.
lesions. Comparison of four chemotherapy regimens for
37. Tumor Ablation for Patients with Lung Cancer 465

advanced non-small-cell lung cancer. N Engl J 24. Vaughn C, Mychaskiw G 2nd, Sewell P. Massive
Med 2002;346:92-98. hemorrhage during radiofrequency ablation of a
13. Breathnach OS, Freidlin B, Conley B, et al. pulmonary neoplasm. Anesth Analg 2002;94:
Twenty-two years of phase III trials for patients 1149-1151.
with advanced non-small-cell lung cancer: 25. Andre F, Grunenwald D, Pujol JL, et al. Patterns
sobering results. J Clin OncoI2001;19:1734-1742. of relapse of N2 nonsmall-cell lung carcinoma
14. Livraghi T, Solbiati L, Meloni MF, Gazelle GS, patients treated with preoperative chemother-
Halpern EF, Goldberg SN. Treatment of focal apy: should prophylactic cranial irradiation be
liver tumors with percutaneous radio-frequency reconsidered? Cancer 2001;91:2394-2400.
ablation: complications encountered in a multi- 26. Vaaler AK, Forrester JM, Lesar M, Edison M,
center study. Radiology 2003;226:441-451. Venzon D, Johnson BE. Obstructive atelectasis
15. Solbiati L, Livraghi T, Goldberg SN, et al. Per- in patients with small cell lung cancer. Incidence
cutaneous radio-frequency ablation of hepatic and response to treatment. Chest 1997;111:115-
metastases from colorectal cancer: long-term re- 120.
sults in 117 patients. Radiology 2001;221:159- 27. Marasso A, Bernardi V, Gai R, et al. Radiofre-
166. quency resection of bronchial tumours in com-
16. Curley SA, Izzo F, Delrio P, et al. Radiofre- bination with cryotherapy: evaluation of a new
quency ablation of unresectable primary and technique. Thorax 1998;53:106-109.
metastatic hepatic malignancies: results in 123 28. Rose SC, Fotoohi M, Levin DL, Harrell JH.
patients. Ann Surg 1999;230:1-8. Cerebral microembolization during radio-
17. Curley SA, Izzo F, Ellis LM, Nicolas Vauthey J, frequency ablation of lung malignancies. J Vase
Vallone P. Radiofrequency ablation of hepato- Intervent RadioI2002;13:1051-1054.
cellular cancer in 110 patients with cirrhosis. Ann 29. Dupuy DE, Zagoria RJ,AkerleyW, Mayo-Smith
Surg 2000;232:381-391. WW, Kavanagh PV, Safran H. Percutaneous
18. Gazelle GS, Goldberg SN, Solbiati L, Livraghi T. radiofrequency ablation of malignancies in the
TUmor ablation with radio-frequency energy. lung. AJR 2000;174:57-59.
Radiology 2000;217:633-646. 30. Nishida T, Inoue K, Kawata Y, et al. Per-
19. Miao Y, Ni Y, Bosmans H, et al. Radiofrequency cutaneous radiofrequency ablation of lung
ablation for eradication of pulmonary tumor in neoplasms: a minimally invasive strategy for
rabbits. J Surg Res 2001;99:265-271. inoperable patients. J Am Coll Surg 2002;195:
20. Goldberg SN, Gazelle GS, Compton CC, Mueller 426-430.
PR, McLoud TC. Radio-frequency tissue abla- 31. Shankar S, vanSonnenberg E, Silverman SG,
tion of VX2 tumor nodules in the rabbit lung. TUncali K, Morrison PRo Management of pneu-
Acad Radiol 1996;3:929-935. mothorax during percutaneous radiofrequency
21. Goldberg SN, Gazelle GS, Compton CC, ablation of a lung tumor: technical note. J Thorac
McLoud TC. Radiofrequency tissue ablation in Imaging 2003;18:106-109.
the rabbit lung: efficacy and complications. Acad 32. Morrison PR, vanSonnenberg E, Shankar S, et al.
RadioI1995;2:776-784. Radiofrequency ablation in the thorax; part 1-
22. Johnson BE. Second lung cancers in patients Experiments in the porcine model. AJR 2005;
after treatment for an initial lung cancer. J Nat! 184:375-380.
Cancer Inst 1998;90:1335-1345. 33. vanSonnenberg E, Shankar S, Morrison PR, et al.
23. Dupuy DE, Mayo-Smith WW, Abbott GF, Radiofrequency ablation in the chest; part 2-
DiPetrillo T. Clinical applications of radio- Initial clinical experience with emphasis on
frequency tumor ablation in the thorax. Radio- adjunctive procedures and multidisciplinary
graphies 2002;22(Spec No.):S259-S269. input. AJR 2005;184:381-390.
38
Ablative Therapies for Gastrointestinal
Malignancies: The Gastrointestinal
Oncologist's Viewpoint
Matthew Kulke

Surgical resection, radiation therapy, and sys- limited hepatic disease. In a large, multiinstitu-
temic chemotherapy are the primary treatment tional French study, over 1800 patients under-
modalities for patients with gastrointestinal going surgical resection for colorectal cancer
malignancies. Ablative therapies such as metastases were found to have a 5-year survival
radiofrequency ablation (RFA) and cryoabla- rate of 25% (1). Other large series have
tion hold potential as alternative treatment reported 5-year survival rates of 25% to 37%
options that, in the appropriate setting, may be for patients undergoing complete resection of
both efficacious and well tolerated. However, liver metastases (2-8).
while ablative therapies are used commonly in Patient selection plays a critical role in deter-
the treatment of gastrointestinal malignancies, mining who may benefit from hepatic resection.
their precise role in such patients is poorly The presence of extrahepatic sites of disease
defined. Initial studies of ablative therapies is a uniformly poor prognostic factor, and is
have focused on malignancies such as colorec- usually a contraindication to hepatic resection
tal cancer, hepatocellular carcinoma, and neu- (2,5,7,8). Exceptions to this general rule include
roendocrine tumors, in which surgical resection, extrahepatic disease limited to a resectable
particularly of metastatic lesions, has played an local recurrence, or potentially resectable pul-
important role. Ablative therapies are less monary metastases. The synchronous presenta-
likely to playa significant role in the treatment tion of hepatic metastases and a primary tumor,
of gastric and pancreatic cancer, which are as compared to the development of hepatic
more commonly associated with widespread metastases following resection of the primary,
dissemination and peritoneal disease. also is an adverse prognostic factor (5,7,8).
Some studies have suggested that the presence
of four or more hepatic metastases is a con-
traindication to surgery. However, a recent
Ablative Therapies in study from Memorial Sloan-Kettering Cancer
Colorectal Carcinoma Center found that patients undergoing com-
plete resection of four or more metastases
Ablative therapies for colorectal cancer usu- experienced a 5-year survival of 24%, suggest-
ally are considered in patients with relatively ing that in properly selected patients, resection
limited hepatic metastases. Interest in the of multiple metastases is potentially beneficial
ablation of hepatic metastases from colorectal (2).
cancer stems from the historical success of Ablative therapies, such as cryoablation or
hepatic resection in this setting. Several series RFA, generally are reserved for patients whose
have firmly established hepatic resection as a liver metastases are not considered surgically
potentially curative procedure in patients with resectable, but may still be amenable to therapy

466
38. Ablative Therapies for Gastrointestinal Malignancies 467

TABLE 38.1. Selected studies of radiofrequency ablation in patients with metastatic colorectal cancer.
Study No. of Patients No. of Lesions Median follow-up (months) Disease-free survival
Rossi. 1996 (32) 11 13 22 1
Rossi, 1998 (33) 14 19 12 2
De Baere. 2000 (9) 68 121 13.7 NA
Solbiati, 2001 (10) 117 179 36 36%

with curative intent. Such scenarios include from 11%-40%, with no studies reporting
location of tumors located close to a blood median follow-up beyond 3 years (Table 38.2).
vessel, or the presence of comorbidities that The potential benefit of ablative therapy as a
may preclude surgery. Studies of RFA in strictly palliative treatment for patients with
patients with metastatic colorectal cancer are hepatic metastases from colorectal cancer has
limited by inconsistent selection criteria and not been studied. Currently, the standard
short follow-up (Table 38.1). In an early Italian palliative approach to patients with unresec-
study, 14 patients with hepatic metastases table metastatic colorectal cancer is systemic
underwent RFA. Of these patients, only two chemotherapy. First-line treatment, utilizing a
patients remained disease-free. In a subsequent combination chemotherapy regimen containing
French study, de Baere and colleagues (9) irinotecan, 5-fluorouracil, and leucovorin, has
treated 68 patients, the majority of whom had clearly been shown to result in palliation of
metastases from colorectal cancer. The initial symptoms and improvement in overall survival
results of this study were encouraging, with suc- (11). Combinations of 5-fluorouracil and
cessful ablation of 91 of 100 treated metastases. oxaliplatin also have activity in patients with
However, the longer-term benefits of these pro- metastatic colorectal cancer. Oxaliplatin is used
cedures were not reported. Long-term results commonly for the treatment of colorectal
from RFA were reported first by Solbiati and cancer in Europe, and was recently approved
colleagues (10), who performed RFA in 117 for use as second-line therapy for metastatic
patients with metastases from colorectal colorectal cancer in the United States (12).
cancer. With a median follow-up time of 3 years, The toxicities associated with systemic
an overall survival rate of 36% was achieved. chemotherapy have led to studies of hepatic-
These results are similar to those achieved directed therapy in patients with extensive
with surgical resection, and warrant a direct hepatic metastases not amenable to surgical
comparison between these two treatment resection. Several studies have evaluated the
modalities. palliative benefit of chemotherapy infused
Interpretation of the results of cryoablation directly into the hepatic artery. This technique
in the treatment of hepatic metastases from provides higher concentrations of chemother-
colorectal cancer similarly is limited by a lack apy directly to the metastatic sites, while limit-
of strict selection criteria and short follow-up. ing systemic toxicity. Hepatic arterial infusion
Disease-free survival rates appear similar to has been shown to result in higher response
those of patients undergoing RFA, and range rates than systemic chemotherapy, resulting in

TABLE 38.2. Selected studies of cryoablation in patients with metastatic


colorectal cancer.
Median follow- Disease-free
Study No. of Patients up (months) survival
Onik. 1993 (12) 69 12-15 40%
Ravikumar, 1991 (34) 24 24 28%
Weaver, 1995 (35) 47 30 11%
468 M. Kulke

TABLE 38.3. Selected studies of radiofrequency ablation in patients with hepatocellular carcinoma.
Study No. of Patients No. of Lesions Median follow-up (months) Disease-free survival
Rossi, 1996 (32) 39 41 22.6 23 (59%)
Ianitti, 2002 (36) 30 NA 36* 3 (60%)*
Kuvshinoff, 2002 (37) 11 NA 9 6 (53%)

* Results from only five patients reported.

shrinkage of liver metastases in approximately supplying the tumor are occluded, is associated
50% of cases (13,14). However, these trials with tumor response rates in excess of 50%.
have failed to demonstrate a meaningful sur- Two studies in which chemoembolization was
vival benefit associated with the hepatic performed in patients prior to liver transplan-
arterial infusion, in large part because of the tation demonstrated encouraging long-term
subsequent development of extrahepatic dis- survival rates (17,18). The long-term benefit of
ease. Based on this experience, it is difficult to chemoembolization in patients who do not
recommend RFA or cryoablation as a strictly undergo transplantation, however, remains
palliative treatment outside of a clinical trial uncertain. Systemic chemotherapy has played
setting. little role in the treatment of patients with unre-
sectable hepatocellular carcinoma. Doxoru-
bicin and 5-fluorouracil-based regimens have
Ablative Therapies in failed to demonstrate significant activity in this
Hepatocellular Carcinoma disease, and do not improve patient survival sig-
nificantly (19,20).
Currently, the only established cure for The limited treatment options available to
hepatocellular carcinoma is partial or total patients with unresectable hepatocellular carci-
hepatectomy with liver transplantation. Partial noma have made newer techniques such as
hepatectomy is curative in over 30% of selected RFA and cryoablation potentially attractive
patients (15,16). The operative mortality asso- alternatives. Initial small studies of RFA in
ciated with partial hepatectomy has decreased patients with hepatocellular carcinoma show
to less than 5% in recent years, in large part due relatively high rates of disease-free survival,
to improved operative technique and better although follow-up times remain short. The
patient selection. Baseline hepatic function is successful use of cryoablation also has been
one of the most important factors that influence reported in patients with hepatocellular carci-
patient outcome. Poor prognostic factors in noma (21,22) (Table 38.3).
patients being considered for hepatic resection
include the presence of cirrhosis, portal hyper-
tension, thrombocytopenia, and coagulopathy Ablative Therapies in
(4). In patients with poor underlying hepatic Neuroendocrine Tumors
function, hepatic transplantation may be con-
sidered. The limited number of livers available The clinical course of patients with metastatic
for transplantation, however, has limited the carcinoid and pancreatic neuroendocrine
utility of hepatic transplantation as a routine tumors is highly variable. Some patients with
treatment for hepatocellular carcinoma. indolent tumors may remain symptom-free for
In patients who are not candidates for years, even without treatment, while others
hepatic resection, or for whom transplantation more clearly require therapy. Several options
is not feasible, other treatment modalities can are available for the treatment of metastatic
be considered, although none has been demon- neuroendocrine tumors. While cytotoxic
strated to be beneficial. Chemoembolization, a therapy has played only a limited role in the
technique in which the hepatic arterial vessels management of patients with metastatic neu-
38. Ablative Therapies for Gastrointestinal Malignancies 469

roendocrine tumors, treatment with somato- lation, either alone or in conjunction with sur-
statin analogues, surgical resection, hepatic gical debulking. Most published reports are
artery embolization, and, more recently, RFA small case studies of fewer than 40 patients
and cryoablation can be considered in appro- (28). In the series with the longest follow-up, 31
priate patients. symptomatic patients with metastatic carcinoid,
Patients with metastatic neuroendocrine islet cell tumor, or medullary thyroid cancer,
tumors often become symptomatic from hor- underwent resection, cryosurgery, and/or RFA
monal hypersecretion rather than from tumor (29). Symptoms were eliminated in 27 (87%),
bulk. This is especially true for patients with and 16 had progressive or recurrent disease
small bowel or appendiceal carcinoid tumors, in with a median follow-up of 26 months.
whom symptoms are typically absent until
hepatic metastases supervene. The secretion of
serotonin and other vasoactive substances into Ablative Therapies in
the systemic circulation results in the carcinoid
syndrome. The carcinoid syndrome is charac-
Gastric Cancer
terized by episodic flushing, wheezing, diarrhea,
Surgical resection is the only potentially cura-
and the eventual development of carcinoid
tive treatment for patients with gastric cancer.
heart disease. Symptoms of the carcinoid syn-
Despite surgical resection, over 50% of patients
drome can be controlled successfully by the
subsequently develop metastatic disease. In
administration of somatostatin analogues in
contrast to colorectal cancer, hepatocellular
approximately 90% of cases (23). Actual radio-
carcinoma, and neuroendocrine tumors, the
logic tumor regression following treatment with
pattern of metastatic spread in patients with
somatostatin analogues is rare, however (24).
gastric cancer often involves the peritoneum.
Resection of hepatic metastases also can
Because of the high incidence of peritoneal
effectively palliate symptoms of hormonal
disease, there is rarely an indication for metas-
hypersecretion, and may, in some cases, result
tatectomy or ablation in such patients.
in long-term survival. In one series of 74
patients undergoing hepatic resection for
metastatic disease, over 90% experienced
symptomatic relief, and the 4-year survival rate Ablative Therapies in
exceeded 70% (25). In general, resection is Pancreatic Cancer
undertaken only in patients with a limited
number of hepatic metastases, and is most suc- Patients with early-stage pancreatic cancer
cessful when undertaken with curative intent. are candidates for pancreaticoduodenectomy
Hepatic artery occlusion or embolization is an (Whipple resection), which is the only poten-
alternative therapy for patients who are not tially curative option for this disease. Unfortu-
candidates for hepatic resection. The response nately, only 15% of patients with pancreatic
rates associated with embolization, as mea- cancer are candidates for surgical resection.
sured either by decrease in hormonal secretion The remaining patients generally present with
or by radiographic regression, generally are locally unresectable disease due to involvement
greater than 50% (26,27). However, the dura- of peripancreatic lymph nodes and the celiac
tion of response can be brief, ranging from 4 to vessels, or with distant metastases. Combina-
24 months in uncontrolled series (27). Further- tions of systemic chemotherapy and external-
more, the potential for treatment-associated beam radiation therapy have been shown to
morbidity, which can include abdominal pain, palliate symptoms effectively and prolong sur-
nausea, elevated liver function tests, and infec- vival in patients with locally advanced disease
tion, needs to be weighed against the potential (30). As with gastric cancer, pancreatic cancer
benefits of this procedure. frequently is associated with peritoneal carci-
Other approaches to the treatment of hepatic nomatosis. Systemic chemotherapy with gem-
metastases include the use of RFA and cryoab- citabine has been shown to improve quality of
470 M. Kulke

life and overall survival in patients with 13. Chang A, Schneider P, Sugarbaker P, Simpson C,
metastatic pancreatic cancer (31). A role for Culnane M, Steinberg S. A prospective random-
ablative therapies in either locally advanced or ized trial of regional versus systemic continuous
metastatic pancreatic cancer, therefore, is 5-fluorodeoxyuridine chemotherapy in the treat-
ment of liver metastases. Ann Surg 1987;206:
uncertain.
685-693.
14. Kemeny N, Daly L, Reichman B, Gellar N, Botet
References J, Oderman P. A randomized study of intrahe-
patic versus systemic infusion of fluorodeoxyuri-
1. Nordlinger B, Guiguet M, Vaillant J, et al. Surgi- dine in patients with liver metastases from
cal resection of colorectal carcinoma metastases colorectal cancer. Ann Intern Med 1987;107:
to the liver: a prognostic scoring system to 459-465.
improve case selection based on 1568 patients. 15. Hanazaki K, Kajikawa S, Shimozawa N, et al.
Cancer 1996;77:1254-1262. Survival and recurrence after hepatic resection
2. Fong Y, Fortner J, Sun R, Brennan M, Blumgart of 386 consecutive patients with hepatocellular
L. Clinical score for predicting recurrence after carcinoma. J Am Coll Surg 2000;191:381-388.
hepatic resection for metastatic colorectal 16. Shimada M, Rikimaru T, Sugimachi K, et al. The
cancer: analysis of 1001 consecutive cases. Ann importance of hepatic resection for hepatocellu-
Surg 1999;230:309-321. lar carcinoma originating from nonfibrotic liver.
3. Hughes K, Rosenstein R, Songerabodi S, et al. JAm Coll Surg 2000;191:531-537.
Resection of the liver for colorectal carcinoma 17. Mazzaferro V, Regalia E, Doci R, et al. Liver
metastases: a multi-institutional study of long- transplantation for the treatment of small hepa-
term survivors. Dis Colon Rectum 1988;31:1-4. tocellular carcinomas in patients with cirrhosis.
4. Jamison R, Donohue J, Nagorney D, Rosen C, N Engl J Med 1996;334:693-699.
Harmsen W, Ilstrup D. Hepatic resection for 18. Venook A, Ferrell L, Roberts J, et al. Liver trans-
metastatic colorectal cancer results in cure for plantation for hepatocellular carcinoma: results
some patients. Arch Surg 1997;132:505-510. with preoperative chemoembolization. Liver
5. Jenkins L, Milikan K, Bines S, Staren E, Doolas Transplant Surg 1995;1:242-248.
A. Hepatic resection for metastatic colorectal 19. Lai C, Wu P, Chan G, Lok A, Lin H. Doxorubicin
cancer. Am Surg 1997;63:605-610. vs. no antitumor therapy in an inoperable hepa-
6. Rees M, Plant G, Bygrave S. Late results justify tocellular carcinoma: a prospective randomized
resection for multiple metastases from colorec- trial. Cancer 1988;62:479-483.
tal cancer. Br J Surg 1997;84:1136-1140. 20. Falkson G, MacIntyre J, Moertel C, Johnson L,
7. Scheele J, Stang R, Alterndorf-Hofmann A, Paul Scherman R. Primary liver cancer: an Eastern
M. Resection of colorectal liver metastases. Cooperative Oncology Group trial. Cancer 1984;
World J Surg 1995;19:59-71. 54:970-977.
8. Wanebo H, Chu Q, Vezeridis M, Soderberg C. 21. Perglozzi J, Auster M, Conaway G, Sardi A.
Patient selection for hepatic resection of colo- Cryosurgery for unresectable primary hepato-
rectal carcinoma metastases. Arch Surg 1996; cellular carcinoma: a case report and review of
131:322-329. the literature. Am Surg 1999;65:402-405.
9. de Baere T, Elias D, Dromain C, et al. Radiofre- 22. Sheen A, Poston G, Sherlock D. Cryotherapeutic
quency ablation of 100 hepatic metastases with ablation of liver tumours. Br J Surg 2002;89:
mean follow-up of more than 1 year. AJR 2000; 1396-1401.
175:1619-1625. 23. Kvols L, Moertel C, O'Connell M, Schutt A,
10. Solbiati L, Livraghi T, Goldberg SN, et al. Per- Rubin J, Hahn R. Treatment of the malignant
cutaneous radio-frequency ablation of hepatic carcinoid syndrome: evaluation of a long-acting
metastases from colorectal cancer: long-term somatostatin analog. N Engl J Med 1986;315:
results in 117 patients. Radiology 2001;221:159- 663-666.
166. 24. Leong W, Pasieka 1. Regression of metastatic
11. Saltz L, Cox J, Blanke C, et al. Irinotecan plus flu- carcinoid tumors with octreotide therapy: two
orouracil and leucovorin for metastatic colorec- case reports and a review of the literature. J Surg
tal cancer. N Engl J Med 2000;343:905-914. OncoI2002;79:180-187.
12. Onik G, Atkinson D, Zemel R, Weaver M. 25. Que F, Nagorney D, Batts K, Linz L, Kvols L.
Cryosurgery of liver cancer. Semin Surg Oncol Hepatic resection for metastatic neuroendocrine
1993;9:309-317. carcinomas. Am J Surg 1995;169:36-42.
38. Ablative Therapies for Gastrointestinal Malignancies 471

26. Ruszniewski P, Rougier P, Roche A, et al. citabine as first-line therapy for patients with
Hepatic arterial chemoembolization in patients advanced pancreas cancer: a randomized trial.
with liver metastases of endocrine tumors. A J Clin Oncol 1997;15:2403-2013.
prospective phase II study in 24 patients. Cancer 32. Rossi S, DiStasi M, Buscarini E, et al. Per-
1993;71:2624-2630. cutaneous RF interstitial thermal ablation in the
27. Moertel C, Johnson C, McKuskic M, et al. The treatment of hepatic cancer. AJR 1996;167:
management of patients with advanced carci- 759-768.
noid tumors and islet cell carcinomas. Ann 33. Rossi S, Buscarini E, Garbagnati F, et al. Percu-
Intern Med 1994;120:302-309. taneous treatment of small hepatic tumors by an
28. Hellman P, Ladjevardi S, Skogseid B, Akerstrom expandable RF needle electrode. AJR 1998;170:
G, Elvin A. Radiofrequency tumor ablation 1015-1022.
using cooled tip for liver metastases of neuroen- 34. Ravikumar T, Kane R, Cady B, Jenkins R, Clouse
docrine tumors. World J Surg 2002;26:1052-1056. M, Jr. A 5-year study of cryosurgery in the treat-
29. Chung M, Pisegna J, Spirt M. Hepatic ment of liver tumors. Arch Surg 1991;126:
cytoreduction followed by a novel long-acting 1520-1523.
somatostatin analog: a paradigm for intractable 35. Weaver M, Atkinson D, Zemel R. Hepatic
neuroendocrine tumors metastatic to the liver. cryosurgery in treating colorectal liver metas-
Surgery 2001;130:954-962. tases. Cancer 1995;76:210-214.
30. GITSG. Treatment of locally unresectable carci- 36. lannitti D, Dupuy D, Mayo-Smith W, Murphy B.
noma of the pancreas: comparison of combined- Hepatic radiofrequency ablation. Arch Surg
modality therapy to chemotherapy alone. J Natl 2002;137:422--427.
Cancer Inst 1988;80:751-755. 37. Kuvshinoff B, Ota D. Radiofrequency ablation of
31. Burris H, Moore M, Andersen J, et al. Improve- liver tumors: influence of technique and tumor
ments in survival and clinical benefit with gem- size. Surgery 2002;132:605--612.
39
Treatment of Hepatocellular
Carcinoma by Internists
Shuichiro Shiina, Takurna Teratani, and Masao Ornata

Hepatocellular carcinoma (HCC) is one of the more, ultrasound examination is chiefly per-
most common malignancies in the world, espe- formed by internists.
cially in Taiwan, Japan, Korea, and China, as In most hospitals in Japan, internists (gas-
well as in sub-Saharan Africa (1). This cancer troenterologists or hepatologists) perform
has been increasing in the United States (2) and ultrasound examination of the abdominal
other countries (3). organs, although radiologists and surgeons also
Treatment of HCC is different from that of have access to the examination. Internists
other solid tumors, in that surgery plays a perform almost all interventional ultrasound
limited role. The resectability rate of this cancer procedures. Image-guided percutaneous tumor
is notably low, due to multiple lesions or under- ablations, such as percutaneous ethanol injec-
lying cirrhosis (4). It is estimated that only 20% tion therapy (PElT), percutaneous microwave
to 30% of patients with HCC can be candidates coagulation therapy (PMCT), and radio-
for hepatic resection. Still worse, the cancer frequency ablation (RFA), are performed
recurs frequently, even after curative surgical chiefly by hepatologists and gastroenterologists.
resection (5,6). Approximately 80% of patients In our hospital, we hepatologists perform almost
who receive curative surgical resection develop all image-guided percutaneous tumor ablation
recurrence within 5 years after surgery, due to procedures for liver tumors.
intrahepatic metastases that cannot be detected In our department, we have treated 90% of
by imaging modalities before resection, and previously untreated patients with HCC by
multicentric carcinogenesis that arises from image-guided percutaneous tumor ablation,
underlying cirrhosis metachronously. Ortho- because of the local curability, minimal effect
topic liver transplantation may be an effective on residual liver function, and easy repeatabil-
treatment in some cases, but shortage of organ ity for recurrence. We have performed PElT
donors has led to a restricted indication for since 1985,PMCTsince 1995, and percutaneous
HCC. Thus, various nonsurgical therapies have RFA since 1999 (Fig. 39.1), with satisfactory
been introduced for HCC. long-term results. This chapter details our
Transcatheter arterial chemoembolization experience.
and image-guided percutaneous tumor ablation
are widely performed for HCC. Unlike in the
United States, in Japan many transcatheter Percutaneous Ethanol
arterial chemoembolizations and most image- Injection Therapy
guided percutaneous tumor ablations have
been performed not by radiologists, but by Introduction of PElT (7) brought about a
internists. This is because internists have access drastic change in the treatment of HCC. It has
to angiography and to ultrasound. Further- enabled us to treat HCC effectively by nonsur-

472
39. Treatment of Hepatocellular Carcinoma by Internists 473

300
35 to 87 years (mean 63 years). Eighty-four
percent had cirrhosis. There was one lesion in
250
357 patients (47%), two in 173 (23%), three in
79 (10%), and four or more in 147 (19%). The
maximum diameter was 1.0 cm or less in 21
patients (3%),1.1 to 2.0cm in 206 (27%),2.1 to
3.0cm in 236 (31 %),3.1 to 5.0cm in 222 (29%),
and 5.1 cm or more in 71 (9%).
We have employed the multiple-needle
insertion technique to perform PElT (16) (Fig.
39.1). By inserting multiple needles and chang-
ing the depth of the needle tips, we can inject
ethanol into several portions in a single treat-
ment session. In some cases, we have also used
PElT with the computed tomography (CT)
FIGURE 39.1. Transition of image-guided percuta- assistance method. In this method, we first
neous tumor ablation for liver tumors at the Depart-
insert needles under ultrasound guidance,
ment of Gastroenterology, University of Tokyo.
There was a drastic shift from percutaneous ethanol and then use CT to confirm that the tip of
injection therapy (PElT) and percutaneous the needle is in the proper site. By using the
microwave coagulation therapy (PMCT) to radio- multiple-needle insertion technique and the
frequency ablation (RFA). Today we perform CT assistance method, we can apply PElT to
ethanol injection only on patients who are allocated lesions more than 5 cm in diameter, and thus we
to ethanol injection because of a randomized con- can treat almost all patients with HCC by PElT.
trolled trial. All other patients are treated by RFA. We perform PElT twice a week, until it is
assumed that ethanol is injected throughout the
lesion. Then, CT is performed to determine
gical measures (8-10). Histopathologic exami- whether or not there is any viable cancer. If
nations after PElT have indicated its capacity there remains any undestroyed portions, PElT
to destroy the tumor completely in most cases is repeated until CT confirms entire tumor
(11), and it has achieved considerably high necrosis; ethanol is injected into possible viable
long-term survival rates (12-14). In Japan, parts until they are destroyed completely.
PElT generally has been accepted as an alter- In cases of three or fewer lesions, we usually
native to surgery for small HCCs (15). perform PElT without combining transcatheter
General requirements we currently utilize arterial embolization (TAE). In cases of four or
for PElT are listed in Table 39.1. more lesions, we first perform TAE and then
Between 1985 and 1998, 849 new patients add PElT for main lesions and/or lesions not
with HCC were admitted to our institution. treated sufficiently by TAB. Among the 756
Among them, 756 patients (89%) were treated patients, PElT alone was performed in 597
by PElT. Ages of the patients ranged from (79%) and TAE was combined in the remain-
ing 159.
With regard to long-term efficacy, the sur-
TABLE 39.1. General requirements for percutaneous vival rates of all 756 patients treated by PElT
ethanol injection therapy were 89% at 1 year, 64% at 3 years, 39% at 5
years, and 18% at 10 years. In 394 patients who
Umesectable lesions or refusal of surgery
Absence of apparent vascular or biliary invasion had three or fewer lesions and all patients
Absence of uncontrollable ascites whose lesions were 3 cm or less in diameter, sur-
Absence of marked bleeding tendency; prothrombin vival was 93% at 1 year, 76% at 3 years, 50% at
times ~35% and platelet count ~40,OOOfmm3 5 years, and 27% at 10 years. Furthermore, in
Serum bilirubin level <4.0mgfdL 594 patients in whom all lesions detected by
Informed consent
imaging modalities underwent PElT for a
474 S. Shiina et al

potentially curative treatment, survival was


93% at 1 year, 72% at 3 years, 47% at 5 years,
and 24 % at 10 years. In the remaining 130
patients, whom we classify into the absolutely
noncurable group, only main lesions were
treated by PElT; some were left untreated by
PElT because of too many lesions.
Major complications that were encountered
included peritoneal bleeding in nine patients,
seeding of cancer cells in eight, hemobilia in
seven, and massive hepatic infarction in three.
There were three procedural deaths.

Percutaneous Microwave
Coagulation Therapy
The microwave delivery system (Microtaze;
Azwell, Osaka, Japan) consists of a generator
(Fig. 39.2) and an electrode (Fig. 39.3). The elec-
trode is connected to the generator by a soft
coaxial cable.
In PMCT, heat destroys the cancer tissue FIGURE 39.3. Microwave electrodes for percuta-
generated by microwave energy emitted from neous liver tumor ablation. (A) The electrodes are
the inserted electrode (17,18). The tissue 1.6 or 2.0mm in diameter and 15 or 25cm in length.
around the electrode is coagulated in a spindle The electrodes consist of a stainless steel needle
shape at the level of the monopolar electrode, (external electrode) with a 1-cm-long monopolar
but not beyond; PMCT typically destroys a electrode (internal electrode) at the tip. (B) Tip of
the electrode. The dista12.0cm is the active radiating
portion. Microwaves are emitted between the exter-
nal and internal electrodes, which resemble magnetic
dipoles; therefore, microwave coagulation proceeds
from the base of the monopolar electrode.

certain amount of tissue, although the necrotic


area it induces is smaller than that with
PElT.
General requirements we currently utilize
for PMCT are listed in Table 39.2. They are
somewhat different from those of PElT, be-
cause a considerably large introducing needle
(14 gauge) is used and one treatment session
FIGURE 39.2. Microwave generator. Microwave gen- tends to be more invasive.
erator is a surgical equipment originally developed
in Japan to realize bloodless surgery particularly for We have performed PMCT on 186 patients
fragile parenchymatous organs, like liver. Microwave with HCC, including the primary lesion in 122
generator emits a 2450-MHz microwave. In the patients, and recurrent lesions in the remaining
treatment of liver tumors, not only percutaneous 64. Among the 122 patients with previously
approach but also laparotomic and laparoscopic untreated HCCs, there was one lesion in 67
approaches are applied. (55%), two in 30 (25%), three in 12 (10%), and
39. Treatment of Hepatocellular Carcinoma by Internists 475

TABLE 39.2. General requirements for percutaneous capsular hematoma that required blood trans-
microwave coagulation therapy. fusion in one each, and pyothorax in one.
Unresectable lesions or refusal of surgery Complications were more common when the
Absence of apparent vascular or biliary invasion number of ablations was high in an attempt to
Absence of uncontrollable ascites destroy all lesions completely in one day.
Absence of marked bleeding tendency; prothrombin
times 2:50% and platelet count 2:50,000/mm3
Serum bilirubin level <2.0mgldL
Lesions in portions where the electrode can be inserted Radiofrequency Ablation
and held safely
Informed consent Radiofrequency ablation recently has been
introduced into the treatment of HCC (20-22).
In Japan, RFA has been widely used clinically
since 1999; more than 700 hospitals perform
four or more in 13 cases (11 %). The maximum RFA.
diameter was 1.0cm or less in 5 patients (4%), With RFA, an electrode is inserted into the
1.1 to 2.0cm in 44 (36%),2.1 to 3.0cm in 36 tumor under image guidance. Then radio-
(30%),3.1 to 5.0cm in 26 (21 %), and 5.1cm or frequency energy is emitted from the exposed
more in 11 (9%); PMCT alone was performed portion of the electrode, which is converted
in 54 patients (44%), combination therapy of into heat and causes necrosis of the tumor.
PElT and PMCT was performed in 51 (42%), Heat may be conducted considerably homo-
and the remaining 17 (14%) received all geneously in all directions; the capsule or septa
three-PElT, PMCT, and TAE. of the hepatic lesion may not prevent heat con-
Under ultrasound (US) guidance, introduc- duction. Thus, therapeutic efficacy seems more
ing needles were inserted first and then the reproducible with RFA.
electrode was introduced coaxially through the General requirements for RFA are equal to
needles. Radiation for dielectric heating was those for PMCT. RFA can be used in many
performed at 65 to 85 W for 60 seconds. We cases in which PMCT is difficult. This is because
created an introducing needle with scales, a a much larger area can be destroyed by a single
stopper for the electrode, and a guide needle ablation, and thus fewer electrode insertions
with scales, so that the electrode can be inserted are needed to accomplish the treatment by
into each portion of the lesion systematically RFA.
(19). We have performed RFA on 636 patients
Enhanced CT scan was utilized in all 122 with HCC. There was one lesion in 258 patients
patients to evaluate therapeutic effect of (41 %), two in 168 (26%), three in 88 (14%),
PMCT. In 110 patients for potentially curative four in 58 (9%), and five or more in 64 (10%).
treatment, CT demonstrated complete necrosis The maximum diameter was 2.0 cm or less in
of the lesion with a safety margin. In the 221 patients (35%),2.1 to 3.0cm in 243 (38%),
remaining 12 patients in whom PMCT was 3.1 to 4.0cm in 108 (17%),4.1 to 5.0cm in 31
used only palliatively, CT showed effective (5%), and 5.1cm or more in 33 (5%). Radio-
mass reduction. With regard to long-term effi- frequency ablation was performed with a cool-
cacy, the survival rates of all 122 patients tip electrode (Radionics, Burlington, MA) in
treated by PMCT were 90% at 1 year, 87% at 629 patients and with an expandable-type elec-
2 years, and 68% at 3 years. Local recurrence of trode (RITA, Mountain View, CA) in the other
the treated lesions was encountered in four seven patients. TWo or three days after it was
cases. assumed that entire tumor was destroyed by
Major complications occurred in nine (7%) RFA, CTwas performed to determine whether
of the 122 patients in whom PMCT was used or not there remained any viable cancer tissue.
for the primary lesion: massive pleural effusion If there were any possible undestroyed por-
requiring drainage in three, liver abscess in tions, RFA was repeated until CT confirmed
three, hemoperitoneum, hemothorax, and sub- necrosis of the entire tumor.
476 S. Shiina et al

Lesions were judged to be treated success-


fully by RFA in 634 (99.7%) of the 636 cases
by the final CT (Figs. 39.4 and 39.5). By using
the artificial pleural effusion technique and
the guide needle method, we could ablate
lesions on the surface of the liver, beneath the
diaphragm, near large vessels, adjacent to other
organs, and those that were detected defini-
tively by CT, but that could not be identified
clearly by ultrasound. In the remaining two
cases, we gave up RFA because a lesion was
not detected by ultrasound and did not seem A
to be able to be approached safely because
of the location of the lesion. All lesions
were judged totally necrotic by the final CT.
The mean number of treatment sessions was
2.3 ± 1.4, and the duration of hospital stay was
14.3 ± 7.8 days.
With a mean follow-up of 16 months, 12
patients (1.9%) developed local recurrence of
the treated lesions. Local recurrences devel-
oped in cases in which the border of the lesion
was poorly defined before the treatment, or
when CT scan slices after the treatment were 8
different from those before the treatment due
to differences in inhaling or exhaling, or due
to body movement when ablation was limited
by advanced liver dysfunction, or when there
were too many lesions (greater than 10). The
local recurrence rate was much lower than
those reported by most other investigators.
This is probably because we repeated RFA
until CT confirmed complete destruction of the
tumors.
Complications occurred in 37 (5.8%) of the
636 cases: liver abscess in eight cases, seeding of c
malignant cells in six cases, portal vein throm-
bus in five cases, intraperitoneal bleeding in FIGURE 39.4. (A) Our first case of RFA. A 61-year-
four cases, skin burn in three cases, transient old woman had received microwave ablation under
jaundice in two cases, transient liver failure due thoracoscopic guidance for a lesion beneath the
to massive hepatic infarction in two cases, diaphragm in S4. She was, however, found to have a
local recurrence a year later. (B) The patient
massive pleural effusion in two cases, and bile
received chemoembolization for the local recur-
peritonitis, septic shock, gastric ulcer due to rence. Then she was referred to our institution for
antibiotics, transient deterioration of liver func- further treatment. There is Lipiodol deposit in the
tion due to hepatic vein injury, and abdominal lesion. (C) Computed tomography (CT) scan taken
wall burn with infection in one case each. Only 2 days after RFA. Single insertion of electrode and
one patient with bile peritonitis underwent 12 minutes ablation achieved a nonenhancing area
emergency surgery. All other patients recov- surrounding the Lipiodol deposit.
A

c
FIGURE 39.5. (A) Our second case of RFA. A 48- RFA again, because there was no sufficient safety
year-old man was found to have a lesion 3.6cm adja- margin near the IVe. (C) CT scan taken after the
cent to the inferior vena cava (IVC). (B) CT scan second RFA. We concluded that safety margin adja-
taken 3 days after the first RFA. There was no appar- cent to the IVC had also been obtained. Thus, we
ent undestroyed portion. However, we performed stopped further treatment.
478 S. Shiina et al

ered with nonsurgical measures. There was no be useful in the treatment of metastatic liver
mortality related to RFA. tumors.

Discussion References
1. Okuda K, OkudaH. Primary liver cell carci-
In Japan, primary liver cancer is the fourth most noma. In: Mcintyre N, Benhamou J-P, Bircher J,
common malignancy and was responsible Rizzetto M, Rodes J, eds. Oxford Textbook of
for 34,311 deaths in 2001. More than 95% of Clinical Hepatology. Vol 2. Oxford, England:
primary liver cancer is HCC. Thus, periodic Oxford University Press, 1991:1019-1052.
examination of high-risk subjects, using 2. EI-Serag HB, Mason AC. Rising incidence of
imaging modalities and tumor markers, are hepatocellular carcinoma in the United States.
common practice in Japan to detect HCC at N Eng! J Med 1999;340:745-750.
an early stage. Patients with hepatic cirrhosis 3. Saracci R, Repetto F. Time trends of primary
liver cancer: indication of increased incidence
or chronic hepatitis and hepatitis B surface
in selected cancer registry populations. J Natl
antigen (HBsAg) carriers are regarded as high- Cancer lnst 1980;65:241-247.
risk subjects to develop HCC. 4. Liver Cancer Study Group of Japan. Survey and
We have treated 90% of new patients with Follow-Up Study of Primary Liver Cancer in
HCC by PElT, PMCT, and RFA, with consid- Japan: Report 14. Kyoto: Shinko-insatsu, 2000.
erable success. 5. Balsells J, Charco R, Lazaro JL, et a1. Resection
Percutaneous ethanol injection therapy can of hepatocellular carcinoma in patients with cir-
destroy a relatively large area by a single abla- rhosis. Br J Surg 1996;83:758-761.
tion. However, it is difficult to predict an extent 6. Gouillat C, Manganas D, Saguier G, Duque-
of an ablated area in PElT, because distribution Campos R, Berard P. Resection of hepatocel-
of injected ethanol is affected a great deal by lular carcinoma in cirrhotic patients: long-term
results of a prospective study. J Am ColI Surg
the capsule and septa of the lesion. In PMCT, a
1999;189:282-290.
certain size of necrosis can be obtained repro- 7. Sugiura N, Takara K, Ohto M, Okuda K, Hirooka
ducibly, because heat is conducted homo- N. Percutaneous intratumoral injection of
geneously in all directions; the capsule or septa ethanol under ultrasound imaging for treatment
of the lesion may not prevent conduction. of small hepatocellular carcinoma. Acta Hepatol
However, the size of necrosis induced is con- Jpn 1983;24:920.
siderably small (1.5cm in diameter and 2cm in 8. Livraghi T, Festi D, Monti F, Salmi A, Vettori C.
length). US-guided percutaneous alcohol injection of
RFA has a similar advantage to PEIT in small hepatic and abdominal tumors. Radiology
necrosing a large volume of tumor in one 1986;161:309-312.
session, and similar to PMCT in destroying a 9. Shiina S, Yasuda H, Muto H, et a1. Percutaneous
ethanol injection in the treatment of liver neo-
certain size of tissue reproducibly. Radio-
plasms. AJR 1987;149:949-952.
frequency energy emitted from the electrode is 10. Sheu JC, Sung JL, Huang GT, et a1. lntratumoral
converted into heat, which reliably causes injection of absolute ethanol under ultrasound
necrosis up to 3 cm in diameter. Therapeutic guidance for the treatment of small hepato-
efficacy is more reproducible with RFA than cellular carcinoma. Hepatogastroenterology
with PElT or PMCT. Thus, RFA requires fewer 1987;34:255-261.
treatment sessions and shorter hospital stay to 11. Shiina S,Tagawa K, Unuma T, et a1. Percutaneous
achieve complete necrosis of lesions. Because ethanol injection therapy for hepatocellular car-
of local curability, minimal effect on residual cinoma.A histopathologic study. Cancer 1991;68:
liver function, and repeat therapy for recur- 1524-1530.
12. Shiina S,Tagawa K, Unuma T, et a1. Percutaneous
rence, image-guided percutaneous tumor abla-
ethanol injection therapy of hepatocellular car-
tion, especially RFA, plays an important
cinoma: analysis of 77 patients. AJR 1990;155:
role in the treatment of HCC; RFA also will 1221-1226.
39. Treatment of Hepatocellular Carcinoma by Internists 479

13. Ebara M, Ohto M, Sugiura N, et al. Percutaneous neous microwave coagulation. AJR 1995;164:
ethanol injection for the treatment of small 1159-1164.
hepatocellular carcinoma. Study of 95 patients. 19. Shiina S, Imamura M, Obi S, et al. Percutaneous
J Gastroenterol Hepatol 1990;5:616-626. microwave coagulation therapy for liver tumors.
14. Livraghi T. Percutaneous ethanol injection of J Microwave Surg 1997;15:65-69.
hepatocellular carcinoma: survival after 3 years 20. Buscarini L, Fornari F, Rossi S. Interstitial
in 70 patients. Ital J Gastroenterol 1992;24:72- radiofrequency hyperthermia in the treatment of
74. small hepatocellular carcinoma: percutaneous
15. Shiina S, Imamura M, Ornata M. Percutaneous US-guidance of electrode needle. In: Anderegg
ethanol injection therapy (PElT) for malignant A, Despland PA, Otto R, Henner H, eds.
liver neoplasms. Semin Intervent RadioI1997;14: Ultraschall-diagnostik 91. Berlin: Springer-
295-303. Verlag, 1992, pp. 218-222.
16. Shiina S, Hata Y, Niwa Y, et al. Multiple-needle 21. Rossi S, Di Stasi M, Buscarini E, et al. Per-
insertion method in percutaneous ethanol injec- cutaneous ultrasound-guided radiofrequency
tion therapy for liver neoplasms. Gastroenterol electrocautery for the treatment of small
Jpn 1991;26:47-50. hepatocellular carcinoma. J Intervent Radiol
17. Seki T, Wakabayashi M, Nakagawa T, et al. 1993;8:97-103.
Ultrasonically guided percutaneous microwave 22. Livraghi T, Goldberg SN, Lazzaroni S, Meloni F,
coagulation therapy for small hepatocellular Solbiati L, Gazelle GS. Small hepatocellular
carcinoma. Cancer 1994;74:817-825. carcinoma: treatment with radio-frequency abla-
18. Murakami R, Yoshimatsu S, Yamashita Y, tion versus ethanol injection. Radiology 1999;
Matsukawa T, Takahashi M, Sagara K. Treatment 210:655-661.
of hepatocellular carcinoma: value of percuta-
40
The Surgeon's Perspective on Hepatic
Radiofrequency Ablation
David A. Iannitti

Surgeons continue to have an important role in as systemic and regional chemotherapy, and the
the management of patients with hepatic malig- outcomes of hepatic resection.
nancies. Throughout the decades, modern sur-
gical techniques have improved outcomes in
patients undergoing hepatic surgery. However, Colorectal Carcinoma
the majority of hepatic malignancies con-
tinue to be unamenable to surgical resection. There are approximately 140,000 new cases of
These patients continue to be challenging for colorectal carcinoma diagnosed in the United
clinicians. Surgeons have carried out hepatic States annually (1). At the time of initial diag-
tumor ablation since the 1970s; therefore, a nosis, 25% of patients have hepatic metastases.
vast experience has been gained in terms of Additionally, 20% to 25% of patients develop
techniques, outcomes, and patient selection for metastatic disease within 5 years of diagnosis.
tumor ablation. Cryosurgical ablation has Therefore, 70,000 patients per year are at risk
been performed for the past several decades, of developing metastatic disease from colorec-
and long-term patient outcome data currently tal carcinoma. Approximately 60% of patients
are available. The experience and outcome with metastatic disease have liver involvement,
with cryoablation can be extrapolated to whereas 20% of patients have liver metastases
more modern modalities of tumor ablation, only. Unfortunately, only 25% of patients with
such as with radiofrequency energy. In the metastatic colorectal carcinoma to the liver are
early 1990s, developments in laparoscopic amenable to surgical resection. Survival is
surgery influenced hepatic surgery. Laparo- dependent on the extent of hepatic disease.
scopic hepatic tumor ablation with cryosurgi- Patients with a solitary hepatic metastasis have
cal and radiofrequency probes is available to a median survival of 25 months, patients with
selected patients today. Therefore, the experi- unilobar multicentric disease have a median
ence surgeons have gained in managing pati- survival of 17 months, and patients with diffuse
ents with hepatic malignancies should help to bilobar metastatic disease have a median sur-
select appropriate patients for hepatic radiofre- vival of 3 to 6 months. Few, if any, patients with
quency ablation (RFA). metastatic colorectal carcinoma to the liver
A number of hepatic malignancies can be untreated will survive 5 years (2-4).
treated with RFA, the most common being Standard chemotherapy agents for colorectal
metastatic colorectal carcinoma and hepatocel- carcinoma are 5-fluorouracil (5-FU) and folinic
lular carcinoma. If one is to consider RFA of acid (leucovorin). These chemotherapy agents
these malignancies, it is important to under- usually are well tolerated by patients; however,
stand the natural history of these diseases, as in the presence of metastatic disease, there is a
well as responses to traditional treatment such low response rate and no significant improve-

480
40. The Surgeon's Perspective 481

ment in median or long-term survival (5,6). outcomes (22-24). However, if the patient does
New chemotherapy agents approved by the undergo hepatic resection for liver-only
Food and Drug Administration (FDA) include metastatic disease with clear surgical margins,
irinotecan (CPT-II) and oxaliplatin. These overall survival at 3,5, and 10 years is approxi-
agents can be used alone, in combination with mately 40%, 30%, and 20%, respectively
each other, or in combination with 5-FU and/or (25,26). The role of adjuvant therapy following
leucovorin (7-10). Oxaliplatin, 5-FU, and leu- hepatic resection generally is indicated; how-
covorin currently appear to be the most effec- ever, sufficient long-term data with current
tive systemic therapy regime for metastatic chemotherapeutic agents are inadequate.
colorectal cancer (11-13). However, long-term Following hepatic resection, approximately
studies still are pending at this time. Investiga- 60% of patients develop recurrent disease
tional agents such as epidermal growth factor within 5 years. The liver is involved in 45 % to
receptor inhibitors and signal transduction 75% of cases, and the liver only is affected in
pathway inhibitors, including tyrosine kinase 40% of patients with recurrent disease. Only
inhibitors, currently are being studied in one third of patients with liver-only recurrent
patients with advanced colorectal metastatic metastatic colorectal cancer are candidates for
disease (14). reresection (27,28). The data for repeat hepatic
Hepatic artery infusion for regional resections for recurrent metastatic colon cancer
chemotherapy of the liver for metastatic colon exist; however, there is a small number of
cancer was popularized in the 1980s. The stan- patients per study. However, reresection has
dard chemotherapy agent for hepatic artery been associated with a median survival of
infusion is 5-fluorodeoxyuridine (FUDR) (15). greater than 30 months, with a 5-year survival
There have been mixed reviews in the literature of 16% to 32% (29,30). This suggests that this
evaluating hepatic artery infusion. There does may be a population of patients who would
appear to be some short-term survival improve- benefit from operative therapy. Since 75% of
ment over systemic chemotherapy (16,17). patients with metastatic colorectal cancer to the
Hepatic artery infusion in combination with liver are not candidates for surgery, treatment
current systemic agents continues to be evalu- options, such as hepatic tumor ablation, may be
ated by clinical trials; long-term data still are preferable in these patients. There are many
pending (18). reasons for patients not to be surgical candi-
Hepatic resection continues to be the most dates, such as multicentric disease, bilobar
effective treatment for metastatic colon cancer disease, local anatomic invasion, juxtaposition
to the liver (19). Hepatic surgery has undergone to the major vascular pedicles or structures,
significant improvement, particularly over the and the presence of advanced cirrhosis or other
past 10 years; operative mortality associated comorbid medical conditions (31). Therefore,
with hepatic resections in major centers is less the indications for patients to undergo hepatic
than 3%. Overall morbidity from these proce- tumor ablation of colorectal metastases include
dures is in the 10% to 15% range (20,21). A unresectable tumors, recurrent hepatic tumors,
number of factors affect long-term survival in and the inability to tolerate surgery.
patients who undergo hepatic resection for
metastatic colon cancer to the liver. These
include the number and distribution of lesions, Hepatocellular Carcinoma
resection margins, size of metastases, synchro-
nous versus metachronous development of Hepatocellular carcinoma is a prevalent world-
metastases, the presence of extrahepatic disease, wide disease with approximately 250,000 to one
the type of resection (wedge versus anatomic million new cases diagnosed per year (32,33).
resection), age, gender, blood loss, blood trans- The incidence in the United States is approxi-
fusion requirement, primary tumor characteris- mately 1.8 per 100,000, and in some countries it
tics, as well as carcinoembryonic antigen (CEA) is as high as 30 per 100,000 (32,34). Hepatocel-
level all have been shown to affect long-term lular carcinoma is associated with cirrhosis in
482 D.A. Iannitti

approximately 80% of patients. In many sive disease or impaired liver function that pre-
patients, viral hepatitis B or C infections also cludes hepatic resection. The operative mortal-
are associated (35-37). Hepatocellular carci- ity for patients undergoing hepatic resection for
noma demonstrates a variable growth rate, with hepatocellular carcinoma is higher than in
a median doubling time of approximately 4 other diseases, approximately 11 %, with a
months. The median survival of all patients range of 0% to 25%. Hepatic resection for
diagnosed with hepatocellular carcinoma in hepatocellular carcinoma is associated with
North America who did not receive treatment 1-,3-, and 5-year survival rates of 82%, 44%,
was approximately 9 months (38). Systemic and 31 %, respectively (46,47). Recurrence of
chemotherapy for hepatocellular carcinoma is the disease is high following hepatic resection,
ineffective. The standard agent is doxorubicin with a cumulative recurrence rate at 5 years of
(Adriamycin), which is associated with cardiac 75 % (47,48). Since the majority of patients with
toxicity and a less than 20% response rate (39). hepatocellular carcinoma are not candidates
Other agents, such as liposomal doxorubicin, for hepatic resection because of the advanced
thalidomide, and octreotide have been used; stage of the tumor and underlying liver disease,
however, these alternative agents are associ- patients with isolated disease may be good can-
ated with low response rates and no proven sur- didates for hepatic RFA.
vival benefit (40--42). Investigational research
for treatment of hepatocellular carcinoma
includes vascular endothelial growth factor
receptor inhibitors, cell surface antibody Historical Development of
therapy, and gene therapy; however, the results Tumor Ablation
of systemic chemotherapy for hepatocellular
carcinoma are poor. The concept of tumor ablation certainly is
Regional chemotherapy with hepatic artery not new, as ablation techniques have been
chemoembolization or infusion does appear described for more than a century. In 1845
to have higher response rates than systemic Arnott described using iced saline to treat
therapy. Typical agents used for chemoem- dermal malignancies. Throughout the mid-
bolization or infusion are doxorubicin, cisplat- 1900s, various cryogens were developed, includ-
inum, mitomycin C, and ethiodized oil ing Freon-12, by Paul in 1942, and liquid
(Lipiodol). These agents can be used in diffuse nitrogen by Arlington in 1950. An insulated
infiltrative disease to regionally perfuse the cryotherapy probe was used for treatment
liver and can be repeated on a regular basis. of hepatic malignancies by Cooper in 1963.
Hypervascular nodular tumors can be infused Cryoablation was a fairly popular technique in
initially, and then followed by selective the 1980s and early 1990s. Cryogens such as
embolization of the feeding artery to the lesion. argon gas can be used for cryosurgical therapy
This treatment can be used in preparation for of liver tumors. Later developments in cryoab-
surgery, for ablation, or as the primary therapy lation systems and probes have allowed
for these tumors (43). Past data have demon- cryotherapy to be carried out percutaneously
strated that there have been highly variable (49).
responses to regional chemotherapy; however, Radiofrequency ablation also has been
current prospective randomized trials suggest present since the early 1900s. In 1912, Cushing
there can be significant tumor response with used radiofrequency energy for destruction of
some improvement in survival with regional central nervous system tumors. This technology
chemotherapy for hepatocellular carcinoma. recently has been improved and modified
Two- and 4-year survivals with treatment for treatment of numerous soft tissue tumors.
average 45% and 13%, respectively (44,45). Animal studies that evaluated tissue tempera-
Surgical resection continues to be the treat- ture tolerance have concluded that normally
ment of choice for hepatocellular carcinoma; perfused tissue can tolerate maximal tempera-
however, the majority of patients have exten- tures of approximately 45°C. Temperatures
40. The Surgeon's Perspective 483

above 50°C are cytotoxic to tumors. As tissue presence of extrahepatic disease is associated
temperatures rise, proteins denature, mem- with poor outcomes and precludes a patient's
branes rupture, and cells dehydrate. Radiofre- undergoing local tumor ablation (28,31,52).
quency energy can raise tissue temperatures to Once the presence of extrahepatic disease has
lOO°C, which results in effective coagulative been ruled out, then the liver is examined thor-
necrosis of tumors. In the 1990s, other thermal oughly with intraoperative ultrasound. Ultra-
sources were developed, such as laser and sound is the most sensitive and accurate means
microwave energy for tumor ablation. In 1998 of assessing the extent, location, size, and char-
the FDA approved RFA for therapy of hepatic acteristics of hepatic tumors (53). When all
tumors (50,51). tumors have been located and characterized,
the ablations are planned and executed. Under
real-time radiographic guidance, the RF probe
is inserted to the deep margin of the tumor
Radiofrequency Ablation without traversing a major vascular portal
pedicle. The ablation is performed according to
Materials and Operative the standards of the particular system that is
Techniques being used. The overall goal is to achieve at
least a I-em margin of coagulative necrosis cir-
Radiofrequency generators have been used cumferentially beyond the periphery of the
extensively for the treatment of hepatic tumors. tumor margin.
There are essentially two types of radiofre- Radiofrequency ablation can be supple-
quency generators that are applicable for mented by vascular inflow occlusion to the
hepatic tumor ablation. These are the liver, known as the Pringle maneuver. This
impedance-based systems (RITA, Radiothera- maneuver can be carried out by open surgical
peutics, Mountain View, CA) and the output- techniques, as well as laparoscopically. Inflow
based systems (Radionics; Burlington, MA). occlusion clearly has demonstrated a decrease
The tumor ablation concept is as follows: a in the time required for ablation, as well as
radiofrequency needle probe is passed into the increase in the zone of thermocoagulation
tumor, and then radiofrequency waves from 460 (54,55). The temperature of the margins of the
to 480 kHz are emitted through the tumor. Local tumor should be evaluated during the time
ion agitation from the current raises the tissue of ablation and should be greater than 50°e.
temperature above the cytotoxic threshold (51). With larger tumors, greater than 3cm in diam-
In the impedance-based systems, the current eter, the practitioner should perform overlap-
is begun at a low level, and as the tissue imped- ping zones of ablation, often with multiple
ance increases, the wattage increases for higher passes of the radiofrequency probe to various
current until the volume of tissue within the margins of the tumor to achieve complete
deployable array reaches up to lOO°e. In the tumor ablation.
output-based system, the local tissue tempera- As with any invasive procedure, there are
ture is cooled to approximately 20°e. The gen- risks associated with hepatic RFA. The overall
erator is run at maximal output. The local morbidity averages 0.5%, major complications
cooling effect maintains low tissue impedance approximately 2%, and minor complications
and therefore maximal current is used at the up to 6%. Various complications have been
outset of the ablation through completion. described from hepatic RFA; these include skin
These RFA systems can be used in open OR burns, thrombocytopenia, renal failure, colonic
laparoscopic surgery, as well as percutaneously perforation, bleeding, bile leak and strictures,
under real-time radiographic guidance. The hepatic abscess, fever, liver dysfunction, seg-
basic technique for ablation is relatively similar. mental infarct, portal venous thrombosis,
When performing an operative ablation, the diaphragmatic paralysis, systemic hemolysis,
abdomen is explored thoroughly, either with pneumothorax, and pacemaker dysfunction
open surgical or laparoscopic techniques. The (56-58).
484 D.A. Iannitti

Outcomes of Hepatic studies have short-term follow-up and are per-


formed in an uncontrolled fashion for a variety
Tumor Ablation of pathologies (58,65-68). Curley et al (55)
reported their experience in 123 patients with
As mentioned previously, hepatic cryoablation 169 tumors. Thirty-nine percent were primary
has been performed since the 1970s. Consider- liver cancers, and 61 % had metastatic disease.
able experience and knowledge have been There were no deaths in the series, and a 2.4%
gained from cryoablation. These lessons can complication rate. With a 15-month follow-up,
and should be applied to RFA. It is clear from there was noted to be a 1.8% local recurrence
surgical resection and open surgical cryoabla- rate, and a 27.6% distant recurrence rate. Jiao
tion that if the patient has extrahepatic et al (69) reported a series of 35 patients, includ-
metastatic disease, survival does not differ sig- ing eight patients with hepatocellular carci-
nificantly whether ablation is carried out or not. noma. Four patients were noted to have a 90%
Therefore, extrahepatic peritoneal metastatic reduction in a-fetoprotein (AFP) level. One
disease is a contraindication to ablation. patient died at 2 months; however, the remain-
There has been considerable experience with der of the patients had no evidence of disease
cryosurgical ablation for metastatic colorectal at a lO-month follow-up. Four of 17 patients
carcinoma to the liver. The majority of treated expired, while at 7.6 months the remainder of
patients generally have been considered to have the patients had no evidence of disease. Solbiati
unresectable metastatic disease for a variety et al (70) reported 50% disease-free survival
of reasons; however, it has been demonstrated at 16.6 months with 2-year surVival of 61.5%
that median survival of patients undergoing in patients with gastrointestinal malignancies.
"complete" cryoablation live approximately 36 Rossi et aI's (68) series of patients with small
months, while patients undergoing "incom- hepatocellular carcinomas had a median sur-
plete" cryoablation have a median survival of 24 vival of 44 months, and a 10% local recurrence
months (59). Outcomes for cryoablation for rate. Francica and Marone (71) evaluated 15
metastatic colorectal carcinoma also correlate patients with cirrhosis who had 20 hepatocellu-
with the patient's preablation CEA level. lar carcinomas; there was a 64% disease-free
Patients with CEA levels greater than 100 survival for treated patients at 1 year. Solbiati
ng/mL have significantly worse survival than et al (72) in a follow-up report in 2001, demon-
patients with CEA levels less than 100nglmL strated overall survival of patients undergoing
(60). Five-year survival following surgical hepatic RFA at 1,2, and 3 years of 93%, 62%,
cryoablation for metastatic colorectal carci- and 41 %, respectively. In our experience of
noma is approximately 25% (60-62); this sur- 123 patients (20-month median follow-up), 52
vival outcome is higher than with systemic or patients were treated for colorectal metastases.
regional chemotherapy. One-, 2-, and 3-year survival rates were 87%,
The largest experience of cryosurgical abla- 77%, and 50%, respectively. In 30 patients with
tion for hepatocellular carcinoma (167 hepatocellular carcinoma with a 21-month
patients) is reported by Zhou et al (63,64) from median follow-up, 1-, 2-, and 3 year survival
China. Eighty-six percent of patients were rates were 92%, 75%, and 60%, respectively
noted to have cirrhosis. Overall survival (58).
reported at 1,3, and 5 years was 73.5%, 47.7%,
and 31.7%, respectively. Survival was improved
in patients who had tumors smaller than 5 em.
The survival for patients undergoing cryoabla- Surgical vs. Percutaneous
tion appears to rival that of patients undergo- Ablation
ing hepatic resection.
Radiofrequency ablation outcomes for Since there are currently a number of delivery
hepatic malignancies have been reported in a mechanisms for RFA, one should consider
number of published studies; however, most whether a patient would benefit from an
40. The Surgeon's Perspective 485

open surgical, laparoscopic, or percutaneous tumors that are juxtaposed to other structures,
approach. Currently the majority of patients such as the stomach, colon, or gallbladder, also
undergoing RFA of hepatic malignancies may benefit from an operative approach, par-
undergo the percutaneous approach. However, ticularly a laparoscopic approach when either
several considerations should be given prior to the gallbladder can be removed or the adjacent
treatment of patients. First is the accuracy of structures can be retracted, while the tumor can
diagnostic imaging for detection of hepatic and be more safely ablated with decreased risk of
extrahepatic tumors. Bilchik et al (73) reported visceral injury.
308 patients with unresectable hepatic tumors.
All patients underwent laparoscopy with
laparoscopic and intraoperative ultrasound. Summary
There was a 12% incidence of occult metasta-
tic extrahepatic peritoneal disease that was not Practitioners carrying out hepatic tumor RFA
appreciated by preoperative diagnostic imaging should apply the lessons learned from hepatic
studies. Additionally, 33 % of patients had addi- surgery. Traditional systemic as well as regional
tional lesions not recognized by preoperative treatments generally are ineffective, are associ-
diagnostic imaging. Jarnagin et al (74) reported ated with significant toxicity, and usually do
a trial of 416 patients with "resectable" hepatic not alter patient survival. However, continued
metastatic colorectal carcinomas. Twenty-one research into newer agents may affect survival.
of these patients were deemed to be unre- Systemic chemotherapy for metastatic colorec-
sectable since 11 % had unrecognized occult tal cancer should be considered strongly in
extrahepatic metastatic disease, and 10% were patients as an adjuvant following tumor abla-
noted to have disease greater than predicted by tion to decrease the risk of recurrence or
preoperative diagnostic imaging. Despite the prolong the time to recurrence. Patients with
most current diagnostic imaging techniques larger hepatocellular carcinomas (>4cm) may
available, approximately 10% to 20% of benefit from preablation hepatic artery
patients will have unrecognized extrahepatic chemoembolization to produce a more effica-
peritoneal metastatic disease. These patients cious tumor ablation. Surgical resection contin-
clearly have been shown not to benefit from ues to be the gold standard for survival;
hepatic tumor ablation. Intraoperative ultra- however, in patients with metastatic colorectal
sound, performed either open or laparoscopi- carcinoma and hepatocellular carcinoma,
cally, demonstrates additional lesions not the majority are not candidates for hepatic
recognized by preoperative diagnostic imaging resection for a variety of reasons. Therefore,
in up to 40% of patients. Therefore, laparo- tumor ablation appears to be the most effective
scopic or surgical exploration with intraopera- treatment for patients with unresectable
tive ultrasound currently is the most accurate disease that is confined to the liver. Open sur-
method to assess patients with hepatic malig- gical, laparoscopic, and percutaneous tech-
nancy. This approach should be carefully con- niques can be carried out; however, careful
sidered when a patient is to undergo a planned patient selection for the approach should be
ablation procedure (75,76). made between the surgeon and the interven-
For practical reasons, the operative tional radiologist.
approaches may be more feasible than a per-
cutaneous approach. Patients with more than
three lesions, may benefit from vascular inflow References
occlusion. Also, lesions high on the dome of the 1. Wingo P, Tong T, Bolden S. Cancer Statistics,
liver can be difficult to access percutaneously. 1995. CA Cancer J, Clin 1995;45:8.
Transthoracic approaches have been reported; 2. Wagner JS, Adson MA, van Heerdon JA, et al.
however, there can be associated with pneu- The natural history of hepatic metastases from
mothoraces and/or diaphragm injury, such as colorectal cancer. A comparison with resective
perforation or paralysis (77). Finally, hepatic treatment. Ann Surg 1984;199:502-508.
486 D.A. Iannitti

3. Fong Y, Blumgart LH. Hepatic colorectal metas- growth factor receptors. Br J Cancer 2001;85:
tases: current status of surgical therapy. Oncol- 584-589.
ogy 1998;12:1489-1498. 15. Chang AE, Schneider PD, Sugerbaker PH, et al.
4. Stangl R, Altendof-Hoffman A, Charnley RM, A prospective randomized trial of regional
et al. Factors influencing the natural history of versus systemic continuous 5-fluorodeoxyuri-
colorectal liver metastases. Lancet 1994;343: dine chemotherapy in the treatment of colorec-
1404-1410. tal liver metastases. Ann Surg 1987;206:685-693.
5. Scheithauer W, Rosen H, Kornkek GV, et al. 16. Rougier P, Laplanche A, Huguier M, et al.
Randomised comparison of combination Hepatic arterial infusion of floxuridine in
chemotherapy plus supportive care in patients patients with liver metastases from colorectal
with metastatic colorectal cancer. BMJ 1993;306: carcinoma: long-term results of a prospective
752-755. randomized trial. J Clin Oncol 1992;10:1112-
6. Moertel CG, Fleming TR, MacDonald JS, et al. 1118.
Levamisole and fluorouracil for adjuvant chemo- 17. Lorenz M, Muller HH. Randomized, multicenter
therapy of resected colon carcinoma. N Engl J trial of fluorouracil plus leucovorin administered
Med 1990;322:352-358. either via hepatic arterial or intravenous infu-
7. Douillard JY, Cunningham D, Roth AD, et al. sion versus fluorodeoxyuridine administered via
Irinotecan combined with fluorouracil compared hepatic arterial infusion in patients with nonre-
with fluorouracil alone as first-line treatment for sectable liver metastases from colorectal carci-
metastatic colorectal cancer: a multicentre ran- noma. J Clin Oncol 2000;18:243-254.
domized trial. Lancet 2000;355:1041-1047. 18. Kemeny N, Gonen M, Sullivan D, et al. Phase I
8. Rougier P, Van Cutsem E, Bajetta E, et al. Ran- study of hepatic arterial infusion of floxuridine
domized trial of irinotecan versus fluorouracil and dexamethasone with systemic irinotecan for
by continuous infusion after fluorouracil failure unresectable hepatic metastases from metastatic
in patients with metastatic colorectal cancer. colon cancer. J Clin Oncol 2001;19:2687-
Lancet 1998;352:1407-1412. 2695.
9. Saltz LB, Cox JV, Blanke C, et al. Irinotecan plus 19. Scheele J, Stangl R, Altendorf-Hofman A.
fluorouracil and leucovorin for metastatic colo- Hepatic metastases from colorectal cancer: im-
rectal cancer. Irinotecan Study Group. N Engl pact of surgical resection on the natural history.
J Med 2000;343:905-915. Br J Surg 1990;77:1241-1246.
10. Cunningham D, Pyrhonen S, James RD, et al. 20. Bradley AL, Chapman WC, Wright JK, et al. Sur-
Randomized trial of irinotecan plus supportive gical experience with hepatic colorectal metas-
care versus supportive care alone after fluo- tasis. Am Surg 1999;65:560-566.
rouracil failure for patients with metastatic colo- 21. Harmon KE, Ryan JAJ, Beihl TR, et al. Benefits
rectal cancer. Lancet 1998;352:1413-1418. and safety of hepatic resection for colorectal
11. de Gramont R, Figer A, Seymore M, et al. metastases. Am J Surg 1999;177:402-404.
Leucovorin and fluorouracil with or without 22. Nagashima I, Oka T, Hamada C, et al.
oxaliplatin as first line treatment for advanced Histopathological prognostic factors influencing
colorectal cancer. J Clin Oncol 2000;18:2938- long-term prognosis after surgical resection for
2947. hepatic metastases from colorectal cancer. Am J
12. Levi F, Zidani R, Vannetzel JM, et al. Ran- Gastroenterol 1999;94:739-743.
domised multicentre trial of chronotherapy with 23. Taylor M, Forster J, Langer B, et al. A study
oxaliplatin, fluorouracil, and folinic acid in of prognostic factors for hepatic resection for
metastatic colorectal cancer. Lancet 1997;350: colorectal metastases. Am J Surg 1997;173:467-
681--686. 471.
13. Giachetti S, Itzhaki M, Gruia G, et al. Long-term 24. Altendorf-Hofmann A, Scheele 1. A critical
survival of patients with unresectable colorectal review of the major indicators of prognosis after
cancer liver metastases following infusional resection of hepatic metastases from colorectal
chemotherapy with 5-fluorouracil, leucovorin, carcinoma. Surg Oncol Clin North Am 2003;
oxaliplatin, and surgery. Ann Oncol 1999;10: 162-192.
663-669. 25. Jamison RL, Donohue JH, Nagorney DM, et al.
14. Shaheen RM, Ahmed SA, Liu W, et al. Inhibited Hepatic resection for metastatic colorectal
growth of colon cancer carcinomatosis by anti- cancer results in cure for some patients. Arch
bodies to vascular endothelial and epidermal Surg 1997;132:505-510.
40. The Surgeon's Perspective 487

26. Ohlsson B, Stenram U, Tranberg KG, et al. carcinoma to orally administered thalidomide.
Resection of colorectal metastases: 25-year Am J Clin Oncol 2000;23:319-321.
experience. World J Surg 1998;22:268-276. 42. Kouroumalis E, Skordilis P, Thermos K, et al.
27. Holm A, Bradley E, Aldrete JS. Hepatic resec- Treatment of hepatocellular carcinoma with
tion of metastases from colorectal carcinoma. octreotide: a randomized controlled trial. Gut
Morbidity, mortality, and pattern of recurrence. 1998;42:442-447.
Ann Surg 1989;209:428-434. 43. Ahrar K, Gupta S. Hepatic artery embolization
28. Fong Y, Fortner J, Sun RL, et al. Clinical score for hepatocellular carcinoma: technique, patient
for predicting recurrence after hepatic resection selection, and outcomes. Surg Oncol Clin North
for metastatic colorectal cancer: analysis of 1001 Am 2003;12:105-126.
consecutive cases. Ann Surg 1999;230:309-318. 44. Group d'Etude et de Traitement du Carcinome
29. Bismuth H, Adam R, Navarro F, et al. Re- Hepatocellulaire. A comparison of ethiodized oil
resection for colorectal liver metastases. Surg chemoembolization and conservative treatment
Oncol Clin North Am 1996;5:353-364. for unresectable hepatocellular carcinoma.
30. Nakamura S, Sakaguchi S, Nishiyama R, et al. N Engl J Med 1995;332:1256--1261.
Aggressive repeat liver resection for hepatic 45. Bruix J, Llovet JM, Castells A, et al. Transarte-
metastases of colorectal carcinoma. Surg Today rial chemoembolization versus symptomatic
1992;22:260-264. treatment in patients with advanced hepatocel-
31. Ekberg H, Tranberg KG, Andersson R, et al. lular carcinoma: results of a randomized con-
Determinants of survival in liver resection for trolled trial in a single institution. Hepatology
colorectal secondaries. Br J Surg 1986;73:727- 1998;27:1578-1583.
731. 46. Nagorney DM, Gigot JF. Primary epithelial
32. EI-Serag HB. Epidemiology of hepatocellular hepatic malignancies: etiology, epidemiology, and
carcinoma. Clin Liver Dis 2001;5:87-107. outcome after subtotal and total hepatic resec-
33. Bosch FX, Ribes J, Borras 1. Epidemiology of tion. Surg Oncol Clin North Am 1996;5:283-300.
primary liver cancer. Semin Liver Dis 1999;19: 47. Fong Y, Sun RL, Jarnigin W, Blumgart LH. An
271-285. analysis of 412 cases of hepatocellular carcinoma
34. EI-Serag HB, Mason AC. Risk factors for the at a Western center. Ann Surg 1999;229:790-
rising rates of primary liver cancer in the United 799.
States. Arch Intern Med 2000;160:3227-3230. 48. Belghiti J, Panis Y, Farges 0, et al. Intrahepatic
35. Chu CM. Natural history of chronic hepatitis B recurrence after resection of hepatocellular
virus in adults with emphasis on the occurrence carcinoma complicating cirrhosis. Ann Surg
of cirrhosis and hepatocellular carcinoma. J Gas- 1991;214:114-117.
troenterol HepatoI2000;15(suppl):E25-E30. 49. Ravikumar TS. Interstitial therapies for liver
36. Colombo M. Hepatitis C virus and hepatocellu- tumors. Surg Oncol Clin North Am 1996;5:365-
lar carcinoma. Semin Liver Dis 1999;19:263-269. 377. .
37. Ikeda K, Saitoh S, Suzuki Y, et al. Disease 50. LeVeen RF. Laser hyperthermia and radiofre-
progression and hepatocellular carcinogenesis in quency ablation of hepatic lesions. Semin Inter-
patients with chronic viral hepatitis: a prospec- vent RadioI1997;14:313-324.
tive observation of 2215 patients. J Hepatol1998; 51. Siperstein A, Gitomirsky A. History and techno-
28:930-938. logical aspects of radiofrequency thermoabla-
38. EI-Serag HB, Mason AC, Key C. Trends in sur- tion. Cancer J 2000;6(suppl):S293-303.
vival of patients with hepatocellular carcinoma 52. Adson MA, van Reerden JA. Adson MR, et al.
between 1977 and 1996 in the United States. Resection of hepatic metastases from colorectal
Hepatology 2001;33:62-65. cancer. Arch Surg 1984;119:647-651.
39. Nerenstone SR, Ihde DC, Freidman MA. Clini- 53. Kane RA, Hughes LA, Cua EJ, et al. The impact
cal trials in primary hepatocellular carcinoma: of intraoperative ultrasonography on surgery for
current status and future directions. Cancer Treat liver neoplasms. J Ultrasound Med 1994;13:1-6.
Rev 1988;15:1-31. 54. de Baere T, Bessoud B, Dromaine C. Percuta-
40. Heneghan MA, O'Grady JG. Liver transplanta- neous radiofrequency ablation of hepatic tumors
tion of malignant liver disease. Ballieres Clin during temporary hepatic venous occlusion.AJR
Gastroenterol 1999;13:575-591. 2002;178:53-59.
41. Patt YZ, Hassan NM, Lozano RD, et al. Durable 55. Curley SA, Izzo F, Delrio P, et al. Radiofre-
clinical response of refractory hepatocellular quency ablation of unresectable primary and
488 D.A. Iannitti

metastatic hepatic malignancies; results of 123 68. Rossi S, Buscarini E, Garbagnati F, et al. Percu-
patients. Ann Surg 1999;230:1-11. taneous treatment of small hepatic tumors by
56. Scaife CL, Curley SA. Complication, local recur- an expandable RF needle electrode. AJR 1998;
rence, and survival rates after radiofrequency 170(4):1015-1022.
ablation for hepatic malignancies. Surg Oncol 69. Jiao LR, Hansen PD, Havlik R, et al. Clinical
Clin North Am 2003;12:243-255. short-term results of radiofrequency ablation in
57. Livraghi T, Goldberg SN, Lazzaroni S, et al. primary and secondary liver tumors. Am J Surg
Hepatocellular carcinoma: radiofrequency abla- 1999;177(4):303-306.
tion of medium and large lesions. Radiology 70. Solbiati L, Ierace T, Goldberg SN, et a!' Percuta-
2000;214:761-768. neous US-guided radiofrequency ablation of
58. Iannitti DA, Dupuy DE, Mayo-Smith WW, et al. liver metastases: treatment and follow-up in 16
Hepatic radiofrequency ablation. Arch Surg patients. Radiology 1997;202:195-203.
2002;137:422--427. 71. Francica G, Marone G. Ultrasound-guided per-
59. Shafir M, Shapiro R, Sung M, et al. Cryoablation cutaneous treatment of hepatocellular carci-
of unresectable malignant liver tumors. Am J noma by radiofrequency hyperthermia with a
Surg 1996;171:27-31. "cooled-tip needle." A preliminary clinical expe-
60. Weaver ML, Ashton JG, Zemel R. Treatment rience. Eur J Ultrasound 1999;9(2):145-153.
of colorectal liver metastases by cryotherapy. 72. Solbiati L, Livraghi T, Goldberg, et al. Percuta-
Semin Surg OncoI1998;14:163-170. neous radiofrequency ablation of hepatic metas-
61. Tanden VR, Harmantas A, Gallinger S. Long- tases from colorectal cancer: long-term results in
term survival after hepatic cryotherapy versus 117 patients. Radiology 2001;221:159-166.
surgical resection for metastatic colorectal carci- 73. Bilchik AJ, Wood TF, Allegra D, et al. Cryosur-
noma: a critical review of the literature. Can J gical ablation and radiofrequency ablation for
Surg 1997;40:175-181. unresectable hepatic malignant neoplasms. Arch
62. Ravikumar TS, Cady B, Jenkins R, et al. A 5-year Surg 2000;135(6):657-662.
study of cryosurgery in the treatment of liver 74. Jarnagin WR, Fong Y, Ky A, et al. Liver resection
tumors. Arch Surg 1991;126:1520-1524. for metastatic colon cancer: assessing the risk of
63. Zhou XD, Tang ZY, Yu yO, et al. The role of occult irresectable disease. J Am Coli Surg 1999;
cryosurgery in the treatment of hepatic cancer: 188(1):33--42.
a report of 113 cases. J Cancer Res Clin Oncol 75. Kokudo N, Bandai Y, Imanishi H, et al. Man-
1993;102:100-102. agement of new hepatic nodules detected by
64. Zhou XD, Tang ZY, Yu yO. Ablative approach intraoperative ultrasonography during hepatic
for primary liver cancer. Surg Oncol Clin North resection for hepatocellular carcinoma. Surgery
Am 1996;5:370-390. 1996;119:634-640.
65. Dupuy DE, Zagoria RJ, Akerley W, Mayo-Smith 76. Lo CM, Lai ECS, Liu CL, et al. Laparoscopy
WW, Kavanaugh PV, Safran H. Percutaneous RF and laparoscopic ultrasound avoid exploratory
ablation of malignancies in the lung. AJR 2000; laparotomy in patients with hepatocellular car-
174:57-60. cinoma. Ann Surg 1998;227:527-532.
66. Jeffrey SS, Birdwell RL, Ikeda DM, et al. 77. Shibata T, Iimuro Y, Ikai I, et al. Percutaneous
Radiofrequency ablation of breast cancer: first radiofrequency ablation therapy after intratho-
report of an emerging technology. Arch Surg racic saline solution infusion for liver tumor in
1999;134:1064-1068. the hepatic dome. J Vase Intervent Radiol 2002;
67. Gervais DA, McGovern FJ, Wood BJ, et al. 13(3):313-315.
Radio-frequency ablation of renal cell carci-
noma: early clinical experience. Radiology 2000;
217:665-672.
41
Radiofrequency Tumor Ablation
in Children
William E. Shiels II and Stephen D. Brown

Historical Perspective malformations and tonsillar hypertrophy in


children (11,12). Recently, RF has been applied
Percutaneous image-guided radiofrequency in utero to treat fetal sacrococcygeal teratomas
(RF) tumor ablation continues to gain momen- as well as to occlude vessels in the umbilical
tum in adult patients as a viable and effective cord and placenta in complications related to
therapeutic option in the treatment of solid monochorionic pregnancies (13-16). Reports of
tumors in a variety of locations including the RF tumor ablations in children have been
skeleton, liver, spleen, kidney, adrenal gland, limited predominantly to the musculoskeletal
and lung (1-6). In all these areas, the basic system and osteoid osteomas (2-4). Treatment
concept of RF tumor ablation is similar: local- of hepatoblastoma in children as young as
ized and contained heat generation systemati- 1 year old has been mentioned briefly in the
cally induces focal coagulative necrosis and cell literature (17). This chapter reviews details of
death. Cytotoxicity is best induced when unique care issues in percutaneous RFA in
regional temperatures reach and maintain 50 0 children with respect to general patient care
to lOO°e. In children, RF tumor ablation has concerns and organ-specific RFA applications.
been used most widely in the treatment of The general aspects of RF physics, tissue reac-
osteoid osteoma. Percutaneous RF ablation tion, cellular dynamics, and instrumentation
(RFA) has fewer indications in the pediatric have been discussed in earlier chapters and are
population, and hence, has been slower to not addressed here.
evolve in the treatment of diseases of the liver,
kidney, lung, and soft tissues.
Application of RF energy in children has
been most widely applied in the form of surgi-
Clinical Applications
cal Bovie electrocautery. It is generally con-
sidered that the nature and character of the
General Procedural Issues
coagulative necrotic reaction of the tissues to Although many adult percutaneous RFA pro-
heating with RFA in infants and children is cedures in organs such as the liver, lung, and
similar to that in adults. Historically, RFA of kidney may be successfully performed in adults
tissue has been performed in children for a with conscious sedation, children are not toler-
variety of conditions. In pediatric cardiac elec- ant of similar pain experiences. Thus, general
trophysiology laboratories, atrial and ventricu- anesthesia is required for most, if not all, RFA
lar ablations using RF have been performed procedures. Even with liberal use of periosteal
safely and effectively for many years (7-10). local anesthesia, patients still react to the pain
Reports in the otolaryngology literature have of RF bone ablation if anesthesia is limited to
described using RFA to treat lingual lymphatic deep sedation. Rosenthal et al (18) recently

489
490 W.E. Shiels II and S.D. Brown

described the pain reaction in patients under- volume to body surface area, the potential
going treatment of osteoid osteoma using exists for greater total body heating effect when
general anesthesia; the pain caused elevations large surface areas are treated with RFA in
in cardiac and respiratory rates as biopsy children. In our experience, total body temper-
needles entered the tumor. In our clinical expe- ature elevation has not been encountered in
rience, pain also can be significant enough to the treatment of focal lesions such as osteoid
cause elevation in blood pressure and heart osteoma or focal liver tumors. On the other
rate when the RFA heat reaches sensitive struc- hand, body core temperature elevation to as
tures such as the pleura during RFA of the lung. high as 40 0 e has been documented when treat-
The use of prophylactic antibiotics is variable ing large metastatic lung lesions, and has
among authors and in specific tissue applica- responded well to the use of a hypothermic
tions. Our experience, and that of other authors blanket. When treating large-volume tumors
(2-4), demonstrates that RFA of osteoid (5- to 8-cm diameter), we routinely place the
osteomas is successfully performed without patient on a hypothermic blanket (Medi-Therm
infectious complications in the absence of II; Gaymar, Orchard Park, NY) prior to begin-
prophylactic antibiotics. Authors vary in the use ning the RFA procedure. The use of a hypother-
of prophylactic antibiotics when treating focal mic blanket as cool as 200 e maintains body
hepatic and renal malignancies (2,5,6,19-21). In temperature at or below 38°C during prolonged
the liver, prophylactic antibiotics may be most RFA treatment of large volume tumors in the
appropriate in clinical situations such as severe chest.
cirrhosis, immunosuppression, concomitant Pain control after treatment is a critical issue
infection, and biliary pathology that entail bile and Benign Bone Lesions in children that
stasis, duct dilation, or earlier biliary bypass requires a plan for each specific organ system
surgery. In a large multicenter experience, and lesion being treated with RFA. In the treat-
hepatic abscess was a complication in 0.3% of ment of osteoid osteoma, intraprocedurallocal
2320 patients in whom 3554 liver lesions were anesthesia is coupled with oral analgesics for
treated with RFA (6). In our limited pediatric effective pain control. In the liver and lung, pain
experience, we administer a single dose of pre- from liver capSUlar and pleural distention
procedural antibiotics when treating focal liver usually requires the administration of intra-
lesions, and, to date, none of our cases has been venous narcotic analgesics for effective pain
complicated by abscess formation or sepsis. management. We have found that pain follow-
The potential for thermal skin burns exists ing RFA is greatest 12 to 24 hours following
with RFA due to heat distribution at the skin treatment in the liver and lung, due to accumu-
site of grounding pads used with monopolar RF lation of edema and subsequent increased abla-
systems. Thermal burns are best avoided with tion volume. The pain of postablation syndrome
the use of large surface area foil grounding usually subsides after a few days, and is well
pads, placed with the longest surface edge managed with narcotic analgesia.
facing the RF electrode, at a distance of 25 to
50cm from the electrode (22). In adults, the
thigh is often recommended as a good location
Osteoid Osteoma and Benign
for placement of four large (100cm 2 each) foil
Bone Lesions
pads. In young children, the thighs may be too Osteoid osteoma treatment is the most
small for placement of the large foil pads. As an common application for RFA in children.
alternative, the buttocks serve well as a large Lesions occur in both the axial and appendicu-
surface area placement site for the foil pads lar skeleton. Osteoid osteoma is a nonneoplas-
when RFA is performed in the upper body and tic localized inflammatory focus of eosinophils
trunk. and histiocytes that causes intense recurrent
Young children present with smaller body bone pain. Prior to RFA of osteoid osteoma,
surface areas than their adult counterparts. treatment regimens included long-term use of
Given the relatively greater ratio of tumor nonsteroidal antiinflammatory drugs and sur-
41. Radiofrequency Tumor Ablation in Children 491

gical or percutaneous resection (2-4). Radio- tered into the periosteum prior to biopsy and
frequency ablation therapy of osteoid osteoma into the treatment site at the completion of RF
can be curative in 90% to 94% of patients with treatment to best provide protracted local
a single treatment, and 100% of patients with anesthesia and begin the pain management
two treatments (3,4). regimen.
In treating children with osteoid osteoma, Our current practice utilizes a 200-W RF
general anesthesia is routinely used for RFA. generator (Radionics, Burlington, MA) with a
Computed tomography is the imaging guidance single needle 17-gauge RF electrode, using
modality of choice. Prior to RFA, biopsy either the 0.5- or 1.0-cm exposed tip, depending
specimens are obtained for histologic confirma- on the size of the treated lesion. After the
tion of the correct diagnosis, or to clarify an biopsy is obtained, the outer cannula of the
alternative diagnosis such as intracortical biopsy needle, then subsequently the RF elec-
chondroma (23). trode are placed into the center of the nidus
The biopsy needle serves as the guide for the and the biopsy cannula is retracted away from
RF electrode. In our experience, a 14-gauge the electrode tip. Depending on the size of the
coaxial bone biopsy needle (Ackerman; Cook, lesion, one to two RFA applications are used,
Bloomington, IN) may function well for the with each application providing heat to 90°C
majority of the cases (Fig. 41.1). We have, on for 4 to 5 minutes. Following RFA, patients are
occasion, experienced difficulty passing a 17- discharged under an oral analgesic regimen for
gauge RF electrode through the coaxial pain control. Pain typically subsides in 2 to 3
14-gauge needle lumen. As an alternative to days. Relief of the osteoid osteoma pain usually
the relatively tight fit of a 14/17-gauge coaxial occurs within 12 to 24 hours. The postproce-
needle/electrode system, an ll-gauge bone dural pain is notably different from the original
biopsy needle (M-1 or M-2; Cook, Blooming- pain of the osteoid osteoma. Our experience
ton, IN) provides excellent torque control for with osteoid osteomas parallels that of other
penetrating dense sclerotic bone, delivers a authors who have used RFA to provide an
generous biopsy specimen for pathologic analy- effective cure without complications, and to
sis, and avoids the potential of tight coupling avoid surgical incisions and long-term con-
friction of the guiding needle and electrode. valescence (2-4). The authors have treated
Bupivacaine local anesthesia can be adminis- benign bone tumors such as chondroblastoma

A B

FIGURE 41.1. A 17-year-old boy with osteoid femoral osteoid osteoma nidus (arrow). (B) CT
osteoma, treated with radiofrequency ablation, and image during radiofrequency ablation (RFA) with
then asymptomatic, with 18 months follow-up. (A) the coaxial biopsy needle retracted and the RF elec-
Computed tomography (CT) image of the right trode in the nidus.
492 W.E. Shiels II and S.D. Brown

A c
FIGURE 41.2. Eleven-year-old male with tibial chon- epiphysis. (B) Fluoroscopic spot film image during
droblastoma treated successfully with RF ablation. RF ablation procedure. (C) Follow-up MR image
(A) Plain radiograph of the left knee showing the with no residual tumor and lack of injury to the artic-
focal lytic chondroblastoma in the proximal tibial ular cartilage.

(Fig. 41.2) with success, avoiding operative reported sporadically (17). In our clinical prac-
damage to articular cartilage and physis. tice, focal liver tumors that have been treated
with RFA include hepatoblastoma, hepatic
adenoma, and metastatic leiomyosarcoma.
Hepatic Tumor Ablation Patients with hepatoblastoma who present the
greatest clinical challenge are those with Beck-
The majority of experience documented in with-Wiedemann syndrome who develop mul-
adult hepatic tumor RFA is in the treatment tiple hepatoblastomas over time. Indeed, these
of hepatocellular carcinoma or metastatic are ideal candidates for RFA, given the risks of
disease from organs such as the colon, lung, repeated surgeries for resection (Fig. 41.3).
and breast (1,2,5,6). Treatment of hepatic Hepatocellular carcinoma in our pediatric
tumors (hepatoblastoma) in children has been patients has presented with multiple lesions
41. Radiofrequency Tumor Ablation in Children 493

D
B
FIGURE 41.3. A 2-year-old boy with Beckwith- water-cooled electrode demonstrates echogenic RF
Wiedemann syndrome and focal hepatoblastoma, coagulation necrosis in the tumor. (D) CT image at
treated with RFA. Pretreatment magnetic resonance I-month follow-up demonstrating complete ablation
imaging (MRI) (A) and ultrasound (B) of the focal of the focal hepatoblastoma.
hepatoblastoma. (C) Sonogram during RFA with

(more than four), precluding effective curative bladder may be performed successfully without
options with RFA. In these patients with large, major complications (24). In RFA treatment of
multifocal tumor burden, either systemic pediatric liver tumors, we use a 200- W RF
chemotherapy or chemoembolization is con- generator with internally cooled electrodes
sidered to be a more appropriate treatment (Radionics; Burlington, MA), either as a single
option. electrode or as a cluster of electrodes for larger
General anesthesia is used for RFA of lesions. Overlapping RFAs are performed, with
hepatic tumors in our patients. Single-dose each site reaching a target temperature of at
prophylactic antibiotics are administered prior least 60°C over 12 minutes.
to the onset of RFA treatment. A percutaneous In the treatment of focal liver tumors, the
approach is used in the majority of hepatic experience in children is currently small, as
tumor RFAs. Certain situations are performed pediatric-unique guidelines are being devel-
more appropriately in an open surgical envi- oped. At the current time, the authors would
ronment, particularly to avoid the potential for apply guidelines similar to those in the adult
thermal damage and subsequent perforation in literature, such that we can expect complete
lesions next to the stomach, colon, gallbladder, tumor ablation in 90% of patients with tumors
or other hollow viscus (2). Recent experience smaller than 2.5 cm in diameter (2,5,25). Tumors
has shown that in a small series of patients, that are 2.5 to 3.5 cm in diameter can be
RFA of hepatic tumors adjacent to the gall- expected to be ablated in 70% to 90% of cases;
494 W.E. Shiels II and S.D. Brown

ablation of tumors that are >3.5 to 5.0cm in tions. Furthermore, RFA appears to potentiate
diameter will be seen in only 50% to 70% of the uniform distribution of alcohol in the
cases. In large tumors (>3.5 em) and in con- tumors, which is a challenge in tumors treated
ditions such as the Beckwith-Wiedemann with only ethanol injection. Benign hepatic
syndrome with recurrent hepatoblastoma, lesions treated with RFA offer an alternative to
palliation using RFA is a reasonable goal if partial hepatectomy (Fig. 41.4).
treatment can prolong life expectancy with
limited systemic symptoms and few minor com-
plications. The volume of effective tumor abla-
tion can be enlarged using hypertonic saline
Lung and Renal Tumor Ablation
tumor pretreatment or combined chemoem- Radiofrequency ablation is a promising mini-
bolization (26,27). Most recently, a promising mally invasive option for treatment of solid
combination therapy technique has been devel- lung malignancies in children and adults (2,29).
oped with intratumoral ethanol injection in Unlike adults, children rarely present with
conjunction with RFA (28). In this technique, primary solid malignancies of the lung. Fur-
tumors that were injected with alcohol imme- thermore, relative contraindications to surgery
diately prior to RFA resulted in consistently for lung tumors such as chronic obstructive
larger areas of ablation than tumors treated pulmonary disease (COPD) are much less fre-
solely with RFA, with no increase in complica- quent in children than in adults. In our clinical

A B

FIGURE 41.4. Thirteen-year-old male with biopsy


proven right hepatic lobe hepatic adenoma, success-
fully treated with RF ablation. (A) Diagnostic MR
image with gadolinium demonstrates the focal right
hepatic lobe lesion, percutaneously biopsy proven to
be hepatic adenoma. (B) Intraprocedural sonogra-
phy demonstrates the RF needle in the early phases
of coagulation necrosis in a series of overlapping
ablations. (C) One year follow-up demonstrating
c retraction of the hepatic scar in the ablation bed.
41. Radiofrequency Tumor Ablation in Children 495

A B

c D

FIGURE 41.5. A 12-year-old boy undergoing pal- metastatic lesion, demonstrating early echogenicity
liative RFA for bilateral pulmonary metastatic during RFA (arrow). (E) Sonogram in a later stage
osteosarcoma. (A,B) CT image of the chest demon- of RFA demonstrating large area of echogenicity
strating bilateral pulmonary metastatic lesions, mea- and tumor coagulation (arrows). (F) Prone CT image
suring 5cm (right) and 8cm (left). (C) Sonogram of during second palliative RFA session demonstrating
the osteosarcoma prior to RFA. (D) Sonogram significant bilateral cavitation during RFA.
during ultrasound (US)-guided RFA of the left
496 W.E. Shiels II and S.D. Brown

experience, palliative RFA is currently, and will literature has shown that RFA of renal lesions
likely remain, the most frequent indication for during renal artery balloon occlusion results in
lung tumor RFA in children. In the lung, our larger effective zones of ablation, but with a
experience is limited to the treatment of higher rate of infarction of normal renal tissue
metastatic osteogenic sarcoma in patients who peripheral to the treated focus (30). Other pedi-
are not operative candidates. atric populations that may benefit from RFA of
Internally cooled RF electrodes are used renal masses include those patients with bilat-
with a 200-W RF generator (Radionics; eral Wilm's tumor or nephroblastomatosis, or
Burlington, MA). Large lesions are treated masses arising in solitary kidneys.
with a three-electrode cluster to achieve
burns reaching a minimum target temperature
of 60°C over 12 minutes. We have used both
ultrasound and computed tomography (CT)
Conclusion
guidance for lung RFA cases, with CT most
Radiofrequency tumor ablation in children is
useful when lung precludes an effective
evolving rapidly and offers effective therapeu-
sonographic window. We have provided pallia-
tic options for a variety of pediatric disorders
tive RFA treatment for metastatic lesions as
(31,32). Radiofrequency ablation is well toler-
large as 8cm (Fig. 41.3). The insulating effect of
ated in children and has few complications. As
lung limits the extension of necrosis into adja-
the pediatric experience grows, new opportuni-
cent tissue. In large bilateral tumor ablations,
ties for interdisciplinary treatment regimens
core body temperature elevations are main-
that include RFA will develop, defining clear
tained at 38° to 39°C with the use of a hypother-
roles for this versatile minimally invasive
mic blanket system (Medi-Therm II; Gaymar,
therapy.
Orchard Park, NY), with blanket cooling
temperatures as low as 20°e. Our initial expe-
rience shows that RFA of pediatric lung lesions
is well tolerated. Postprocedural pleuritic pain
References
and nonproductive cough at 12 to 24 hours 1. Goldberg SN, Dupuy DE. Image-guided
may occur, and usually last 2 to 3 days and are radiofrequency tumor ablation: challenges and
well managed with narcotic analgesia. Post- opportunities-part I. J Vasc Intervent Radiol
procedure fever may develop after 24 hours 2001;12:1021-1032.
and lasts 2 to 3 days; blood cultures typically 2. Dupuy DE, Goldberg SN. Image-guided
are negative. In palliative treatment of metasta- radiofrequency tumor ablation: challenges and
opportunities-part II. J Vasc Intervent Radiol
tic lung lesions, the tissue most resistant to 2001;12:1135-1148.
ablation is adjacent to the heart and rib cage, 3. Rosenthal DI, Hornicek FJ, Wolfe MW, Jennings
likely due to the heat-sink effect in these LC, Gephart MC, Mankin HI Changes in the
two areas. Follow-up CT scans may demon- management of osteoid osteoma. J Bone Joint
strate cavitation of the lesion without Surg 1998;80:815-821.
pneumothoraces. 4. Woertler K, Vestring T, Boettner F, Winkelmann
Early experience with percutaneous RFA of W, Heindel W, Lindner N. Osteoid osteoma:
solid renal masses is promising, with renal cell CT-guided percutaneous radiofrequency abla-
carcinoma associated with von Hippel-Lindau tion and follow-up in 47 patients. J Vase Inter-
disease being the most frequent indication in vent RadioI2001;12:717-722.
children and young adults (19-21). In this expe- 5. Ahmed M, Goldberg SN. Thermal ablation
therapy for hepatocellular carcinoma. J Vasc
rience, percutaneous RFA is well tolerated and
Intervent RadioI2002;13:S231-S243.
provides an effective nephron-sparing alterna- 6. Livraghi T, Solbiati L, Meloni MF, Gazelle GS,
tive to surgery. This is particularly important Halpern EF, Goldberg SN. Treatment of focal
with conditions such as von Hippel-Lindau liver tumors with percutaneous radiofrequency
disease, in which patients are predisposed to ablation: complications encountered in a multi-
multifocal renal cell carcinoma. The recent center study. Radiology 2003;226:441-451.
41. Radiofrequency Tumor Ablation in Children 497

7. Campbell RM, Strieper MJ, Frias PA, Danford 21. Roy-Choudhury SH, Cast JE, Cooksey G, Puri S,
DA, Kugler JD. Current status of radiofrequency Breen DJ. Early experience with radiofrequency
ablation for common pediatric supraventricular ablation of small solid renal masses. AIR 2003;
tachycardias. J Pediatr 2002;140:150-155. 180:1055-1061.
8. Hsieh IC, Yeh SJ, Wen MS, Wang CC, Lin FC, Wu 22. Goldberg SN, Solbiati L, Halpern EF, Gazelle
D. Radiofrequency ablation for supraventricular GS. Variables affecting proper system grounding
and ventricular tachycardia in young patients. Int for radiofrequency ablation in an animal model.
J CardioI1996;54:33-40. J Vasc Intervent RadioI2000;11:1069-1075.
9. Manolis AS, Vassilikos V, Maounis TN, 23. Ramnath RR, Rosenthal DI, Cates J, Gebhardt
Chiladakis J, Cokkinos DY. Radiofrequency M, Quinn RH. Intracortical chondroma simulat-
ablation in pediatric and adult patients: compar- ing osteoid osteoma treated by radiofrequency.
ative results. J Intervent Cardiol Electrophysiol Skeletal Radiol 2002;31(10):597--602.
2001;5:443-453. 24. Chopra S, Dodd GD, Chanin MP, Chintapalli
10. Van Hare GF, Dubin AM, Collins KK. Invasive KN. Radiofrequency ablation of hepatic tumors
electrophysiology in children: state of the art. adjacent to the gallbladder: feasibility and safety.
J Electrocardiol2002;35 (suppl):165-174. AIR 2003;180:697-701.
11. Cable BB, Mair EA. Radiofrequency ablation of 25. Livraghi T, Lazzaroni S, Meloni ME Radiofre-
lymphangiomatous macroglossia. Laryngoscope quency thermal ablation of hepatocellular carci-
2001;111:1859-1861. noma. Eur J Ultrasound 2001;13:159-166.
12. Plant RL. Radiofrequency treatment of tonsillar 26. Ahmed M, Lobo SM, Weinstein J, et al.
hypertrophy. Laryngoscope 2002;112:20-22. Improved coagulation with saline solution pre-
13. Tsao K, Feldstein VA, Albanese CT, et al. treatment during radiofrequency tumor ablation
Selective reduction of a cardiac twin by radio- in a canine model. J Vasc Intervent Radiol 2002;
frequency ablation. Am J Obstet Gynecol 2002; 13:717-724.
187:635--640. 27. Bloomston M, Binitie 0, Fraiji E, et al. Trans-
14. Sydorak RM, Feldstein V, Machin G, et al. Feto- catheter arterial chemoembolization with or
scopic treatment for discordant twins. J Pediatr without radiofrequency ablation in the manage-
Surg 2002;37:1736-1739. ment of patients with advanced hepatic malig-
15. Lam YH, Tang MH, Shek TW. Thermocoagula- nancy. Am Surg 2002;68:827-831.
tion of fetal sacrococcygeal teratoma. Prenat 28. Shankar S, vanSonnenberg E, Morrison PR,
Diagn 2002;22:99-101. Tuncali KT, Silverman So. Combined radio-
16. Paek BW, Jennings RW, Harrison MR, et al. frequency and direct alcohol infusion for per-
Radiofrequency ablation of human fetal sacro- cutaneous tumor ablation. AJR 2004;183:1425-
coccygeal teratoma. Am J Obstet Gynecol 2001; 1429.
184:503-507. 29. Dupuy DE, Zagoria RI, Akerley W, Mayo-Smith
17. Iannitti DA, Dupuy DE, Mayo-Smith WW, WM, Kavanagh PV, Safran H. Percutaneous
Murphy B. Hepatic radiofrequency ablation. radiofrequency ablation of malignancies in the
Arch Surg 2002;137:422-426; discussion 427. lung. AJR 2000;174:57-59.
18. Rosenthal DI, Marota II, Hornicek FJ. Osteoid 30. Kariya Z, Yamakado K, Nakatuka A, Onaoda M,
osteoma: elevation of cardiac and respiratory Kobayasi S, Takeda K. Radiofrequency ablation
rates at biopsy needle entry into tumor in 10 with and without balloon occlusion of the renal
patients. Radiology 2003;226:125-128. artery: an experimental study in porcine kidneys.
19. Gervais DA, McGovern FJ, Arellano RS, J Vasc Intervent RadioI2003;14:241-245.
McDougal WS, Mueller PRo Renal cell carci- 31. Hoffer FA. Pediatric applications of radio-
noma: cliniical experience and technical success frequency ablation. Sem Interv Radiol 2003;20:
with radiofrequency ablation of 42 tumors. 323-331.
Radiology 2003;226:417-424. 32. Hoffer FA. Biopsy, needle localization, and radio-
20. Gervais DA, McGovern FJ, Wood BJ, et al. frequency ablation for pediatric patients. Tech
Radiofrequency ablation of renal cell carcinoma: Vasc Interv RadioI2004;6:192-196.
early clinical experience. Radiology 2000;217:
665-672.
42
Comments from Patients and
Their Families
Eric vanSonnenberg

Introduction mailed in. I have scheduled you for a


colonoscopy." How can that be? I had a sig-
To round out the comprehensive ablation story, moidoscopy only three years earlier. I had
we thought who better to tell it than the watched the monitor and heard the doctor say,
patients and families themselves. Finding the "It's clean and pink. Looks good." I had every
initial few patients whom we broached the idea right to think that the sigmoidoscopy gave me
to contribute to be receptive, if not enthusias- a clean bill of health, didn't I?
tic, we decided to include a whole chapter I went in for the colonoscopy that was sched-
devoted to this perspective on ablation. Thus uled. I was nervous. Was blood in the stool
included are the views on ablation from six serious? Again, I chose to watch the monitor. I
patients (with family input). We asked the saw the probe stop and I could see that there
patients to be candid, and edited and redacted was "something" that didn't seem to belong
only names to ensure privacy. there. I asked the doctor if he found something.
The answer was, "Yes, but you are not in a con-
dition right now for me to discuss it with you.
Patient: RR We will wait a little while until the sedative
wears off. Then we will review our findings."
Dr. vanSonnenberg asked me if I, as his patient, Later my wife and I and the doctors sat down
would contribute my thoughts in this text. I was to discuss the findings. The doctor said to me,
very pleased that he asked. I feel that the abla- "You have colon cancer." I felt as though I was
tion procedure, a minimally invasive treatment, hit in the face with a sledgehammer! "We
has certainly extended my life. Of course, I suggest that you consider surgery very soon."
cannot be considered in remission for five more Another sledgehammer! Now I was scared! The
years. But, according to what I have learned, I surgeon explained what needed to be done. Evi-
have a reasonable possibility of living a com- dently, she had watched the monitor during the
plete life span. procedure. She said that the tumor had pene-
I am a male patient at Dana-Farber Cancer trated the colon wall, and that several of the
Institute, 73 years of age, generally in good lymph nodes were involved. Another sledge-
health, slightly overweight, and I exercise hammer! The news got worse with every passing
almost every day. Incidentally, my father, at this moment. Why me? I eat the proper foods. I work
writing, is still alive at 101 years of age. I'm out at the gym almost every day. Why me?
trying to catch up with him. We scheduled the surgery right then. I was
It started when I received a phone call from scheduled for the next day. Instead of going
my primary care physician. He said: "Bob, there home I went to Pre-Op for preparation. She
was blood in two of the fecal samples that you operated the next morning.

498
42. Comments from Patients and Their Families 499

More bad news the next day, after the colec- tumors. One of the methods was called abla-
tomy. She told me that there were indications tion. My friend, Stu, and I spoke two or three
that the cancer had spread to my liver. times a week and compared notes. He provided
However, before the liver surgery was sched- me with a paper from Imaging and Therapeutic
uled, a chest x-ray showed a series of spots on Technology (RSNA, RG volume 20, number 1,
my right lung. More testing was not conclusive. January-February 2000). The information was
So, thoracic surgery was scheduled for August. from numerous national and international
I was scheduled for the liver surgery one universities. It gave me a little insight as to
month later. Please note that I glossed over a what Dr. Eric vanSonnenberg does. Not being
rather difficult recovery due to an infection in, a physician, there were many technical words
or around, the colon. I could not have food so and phrases that I did not understand in the
I was fed intravenously. Needle biopsies and paper. But I did get a little understanding about
numerous other tests were done to determine the procedure and it was apparently low risk.
the cause of the infection. After a while, I was However, I did know that the most effective
given some light food. That turned out to be a way to eliminate cancer tumors was to surgi-
mistake and they had to pump out my stomach. cally remove them. That was confirmed in the
Not too pleasant. paper. But, only a percentage of cancers could
I was scheduled for surgery on the liver at the be removed surgically. This, also, was confirmed
appropriate time. A positron emission tomog- in the text. The paper said that in only one in
raphy (PET) scan indicated that there were three cases would ablation be feasible. I was
three tumors in the liver. My wife told me after- the one in three. I was very scared, but I felt
ward that she thought that everything had gone that having the ablation done could not be any
well, because the surgical procedure lasted over worse than doing nothing other than
three hours. Unfortunately, that was not the chemotherapy.
case. Finally the day came. I went into the hospi-
The liver surgeon spoke to my wife when he tal still not really knowing what to expect. I had
came out of the operating amphitheater. He recent memories of the difficulty after the liver
told her that he could not do anything for me. surgery. I had trouble sitting up because the
He discovered that two of the tumors were too muscles had been surgically cut. It took a while
close to major blood vessels and that another before they were restored. What would be the
one of the tumors was too small to locate. He results of this new ablation procedure? Will it
told us that if he accidentally cut one of the cure my cancer? What are the aftereffects?
blood vessels it would be very serious and What are the risks?
potentially fatal. My wife then asked why the My first ablation procedure was done on
operation had taken so long. The surgeon told October 31,2000. When I woke, in the recovery
us that, during the procedure, he had called in room, surprisingly, I felt great! No pain! I was
two other surgeons to ascertain if they had any told that I would spend the night in the hospi-
suggestions for removing the tumors. They tal. However, the next day, after a test and a
didn't. computed tomography (CT) scan, I could go
My oncologist told me that the chemotherapy home. It sounded too good to be true. It was
was controlling the cancer cells and that they true. I went home the next day and I went back
had shrunk almost in half. But to me to work in a few days. I felt fine. No pain. Minor
the future looked bleak. Very bleak. Then he bandage. To me it was incredible. At one time
told me that there was a new technique. Sign me my oncologist said that they were constantly
up! It sounded like a potential alternative to an working on new technology-something like
almost certain slow and possibly painful end. Star Wars.
My closest friend was diagnosed with colon It would be some months before we would
cancer a few weeks before I was. His brother know the results of that ablation. The burns
and his daughter were both physicians. They would have to heal before the PET scan and
researched various techniques used to destroy other tests could be done.
500 E. vanSonnenberg

In May 2001, a PET scan and CT scan could not take a complete circumferential cuff
detected a tumor in my right lung. The new because of the proximity of a major blood
technology, the PET scan, discovered the very vessel. An extensive needle biopsy was per-
small tumor in my lung. Dr. vanSonnenberg formed, I assume, to confirm the existence of
contemplated using the ablation technique on the cells and to verify the type.
the lung. However, he considered the risks and So, a second ablation procedure was sched-
decided that surgery would be a safer proce- uled for August 22, 2002. From this patient's
dure. I assumed that there was not enough point of view my trepidation was significantly
information or experience to proceed with an lower. I definitely felt more confident. My
ablation on the lung. experiences with surgery versus the ablation
On June 15,2001, the second thoracic surgery procedure left me with a great deal of confi-
was performed. That, also, had limited success. dence in the ablation technique. My confidence
Again, a surgeon was not able to get the entire was justified. The recovery was the same as
cuff around the cell. So, I experienced a month before.... a "piece of cake."
of radiation on the lung. This was to complete When I spoke with Dr. vanSonnenberg after-
the attack on the cuff. ward, he said that he was not positive that he
In September of 2001, I was surveyed, again, was able to ablate a sufficient cuff around the
by a PET scan and a CT scan. I was told that small tumor. He said that it was extremely close
there was a possibility that the tumors had to my diaphragm. There could be serious results
re-appeared. But, Dr. Fuchs, my oncologist if it was damaged. I asked the doctor what
reassured me, that one cell was the one that would happen if a tumor reappeared at that
they originally could not see and the other site. He said, simply: "We'll go in again." That
was a recurrence at the site of one that had did not frighten me at all.
been ablated. Dr. vanSonnenberg, at that time,

Patient and Wife: DM and BM ful demeanor. However, his surgery was con-
sidered successful, and I made the incorrect
assumption that any future cancers would occur
The Patient's Wife
in his colon and could easily be removed, much
I am the patient's wife. We have been married as the first had been. I knew about the statistics
since 1975, and our son, Jonathan was born in of metastases, but the percentages given to us
1983. I was 37 when our son was born, and my for recurrence were comfortably low, especially
husband was 40. I am now 56. I am an architect for a cancer that had been identified in its
for the U.S. Courts, reviewing large projects and early stages, as his had. I know now that the
designing smaller ones. It is an ideal job statistics are skewed, because metastases occur
for me, and I hope to continue working at it much more frequently in men than in women.
for at least another 10 years. Our son has I think that oncologists should tell their
just started college, at Cornell University, and patients this.
we will be faced with large expenses for some When we went to Europe in 1997, we already
time. had indications that something was amiss: his
I have always considered my husband the carcinoembryonic antigen (CEA) levels had
healthy spouse: not overweight, athletic, with been rising. Our oncologist was very watchful,
much physical energy. Therefore, it was a ter- and when the recurrence was confirmed in
rific shock when he was initially diagnosed with 1998, she started him immediately on standard
colon cancer in 1995. We were in the process of chemotherapy. Oddly enough, the physician we
interviewing prep schools for our son, and it consulted with for a second opinion at the
was very difficult to maintain a calm and cheer- Dana-Farber Cancer Institute said that she
42. Comments from Patients and Their Families 501

believed in starting treatments only when the mlstlc (at least outwardly), and sent him to
cancer became symptomatic. It is difficult for Dana-Farber to speak with Dr. vanSonnenberg
laypersons to know which approach to choose. and Dr. Enzinger. Dr. Enzinger recommended
There does seem to be an enormous burden on that he have the ablation and then return to
the patient and family to do their own research him for discussion of other treatments.
and plot their own treatment; this has so far It was in the time span during which the first
worked for us, as we are well-educated people, ablation was delayed, due to an insurance
living in the Boston area (which has major nightmare, that we began reading about the
medical institutions), and we have Internet drug C-225 in newspapers and the popular
access. But even we do not know if we have magazine Science News. We telephoned our
access to all available information. primary oncologist, and asked if she thought
I found out about the results of the con- he would qualify for the upcoming trials to be
clusive CT scan when I was at a meeting in held in Boston and several other cities in the
Providence, R.I. I have no idea how I got home U.S. She verified that he would qualify with
that afternoon. I screamed for some time, and Dr. Enzinger. Dr. Enzinger cheered us along as
my husband comforted me, then somehow we my husband went through a trial of C-225, and
got into the car to pick up our son at summer apparently had above-average positive results.
camp. I cried on and off for several weeks, but It is important here to note that it was my
the human spirit is very resilient. Soon, life husband and I who approached the doctor with
developed a new routine-treatments and questions about the availability of such a trial;
recovering from treatments. But the grief did no one suggested it to us. As I have said, we had
affect me at work; one day, after a shouting an insurance nightmare during which a lengthy
match with a colleague, I called my doctor and battle ensued, which is described below, but the
headed over to her office. She prescribed upshot was that the local BCBS (Federal
Zoloft, clonazepam, and trazodone. I think Employees Service) had not even approached
these drugs have saved my life. the government agency, OPM, that rules
The first approach to controlling the metas- on allowed procedures, but had assumed
tases was radiation therapy; it produced good it was experimental. My husband wrote a bril-
effects, and most of all it was successful, in my liant letter, with the help of his physicians,
mind, because it introduced us to Dr. Anthony proving that it was not at all experimental. It is
Howes, who spoke honestly but optimisti- a terrible health insurance system that is in
cally of all the research and treatments place in this country, and it is heartbreaking
that were coming online. The first round of that it punishes the people with the least
chemotherapy, 5-fluorouracil (5-FU) with leu- amount of spare energy to fight for their
covorin, produced few bad side effects, and life treatment.
continued as usual. The second drug, irinotecan, Now we are considering phase I trials, but my
was more invasive, causing hair loss, diarrhea, hopes are with continued radiofrequency abla-
and the inability to taste food. Each time, we tions (RFAs). He is healthy and looks as good
were given no idea how long the positive effects as he ever has; everyone comments on it. I like
of the treatment would be. Whether this was to think that our love and my ability to make
done so as not to scare us, or because different him laugh will keep him alive. I have never
patients react differently, we did not learn. My loved him more.
husband kept working as an architect through- I have not spoken about our son and his deal-
out this ordeal, arranging his treatments for ings with his father's potentially fatal illness. I
Friday afternoons so that he could go home know it has affected him deeply, and he has
immediately afterward. I went with him to the been able to work some of that out in his
first few, but soon it became routine, and we writing and his art. He argues with his father,
both felt better about letting him go alone. which is normal, and I resist the strong impulse
When the second round of chemo stopped to tell him that he may regret his harsh words
being effective, his oncologist remained opti- sooner than he thinks.
502 E. vanSonnenberg

The Patient the procedure, and I listened as Peter, who was


unaware that I had cancer, described some of
I asked myself, "Why, when Dr. vanSonnenberg his work to another guest. Later, my oncologist
asked me to discuss the goals I had for the informed me that the conventional chemother-
ablation, did I sit dumbfounded?" It was early apy drugs I had been receiving were no longer
August 2002, one day after my 59th birthday, working. In the brief time that elapsed between
and two weeks after I had left the Brigham and this disappointing news and her telling me
Women's Hospital, mostly recovered from my about RFA, I had begun to feel the grip of
second radiofrequency ablation, this time for despair. She mentioned another patient who
two large tumors in my liver. Was it not obvious was helped by RFA. Helped how much? I
to everyone that I wanted to continue living? didn't ask. I knew it wouldn't necessarily be the
Was this not the goal? Or did I not answer same for me, and, should it have been the case,
because I felt it too presumptuous to have I didn't want to hear any stories about how the
goals? Maybe I could have adopted the posture operation had been successful but the patient
of the boardroom executive and announced, had died. I telephoned Peter to inform him that
"Yes! By 2003 we want to be tumor free!" I would be having an appointment with Dr. van-
A more truthful explanation might be that I Sonnenberg, who he then informed me was an
simply did not think of a specific goal for my old friend. Even though Dr. Mueller, by this
ablation. I have metastases from colon cancer, time, had elucidated some of the uses for RFA,
which first became evident two years after the the idea of using high-frequency radio waves
surgery that removed the original tumor and a still seemed fantastic to me and I went to my
section of my colon. This was, now, almost seven first meeting with Dr. vanSonnenberg and his
years ago. After four years of chemotherapy, assistant, Dr. Shankar, in awe, thinking that if
perhaps I had become too suspicious of goals. I were a candidate for this procedure, it would
Maybe my family and I had been through the be my last hope. They explained in detail
success/failure or happiness/sadness cycle of the process and equipment used, the risks,
chemotherapy so many times that the confi- the preparation required, an estimate of the
dence to set goals eluded me, because so often amount of total tumor they thought could be
one can only wait for the next CT scan to see if killed, and the recovery. Immediately I thought
treatment has had any effect on the tumors. there would be tumor remaining that could
One is grateful if one can make it from CT scan continue to grow, but felt reassured to hear that
to CT scan without suffering greatly from some the process could be repeated. The thought of
of the side effects of the drugs. In this way, the risks was sobering, partly because of the
one gradually loses one's sense of reality, or location of the tumors in my specific case.
the awareness and alertness that comes with However, their track record was excellent, and
knowing that one is actually in a continual I felt I needed to have confidence in their expe-
battle with a terrorist. So, I simply muttered rience. I wanted this to work and did not think
something about how I didn't know how suc- twice about doing it due to the risks.
cessful the procedure had been, as if I could The night before my first ablation was
have established a goal after the fact, and in my to take place Dr. vanSonnenberg's assistant,
shyness, like the student who has not done his Dr. Shankar, telephoned me to say that our
homework, blurted out something that might insurance company had denied coverage of the
have been consistent with the results. operation. He was hopeful that the situation
Initially I had learned of RFA casually, while would reverse itself and indicated that this
attending a seasonal cocktail party at the home would most likely happen once the insurance
of Dr. Peter Mueller and his wife Dr. Susan company had received further clarification
Mueller with whom my wife and I had become from the hospital. Surely, it would be resolved
friends, since our sons were classmates and by the end of the week. I became depressed,
baseball teammates in high school. At this time, and during subsequent calls from the doctor
I didn't know I would be a future candidate for later in the week, I remained motionless in the
42. Comments from Patients and Their Families 503

lounge chair while he and my wife discussed the feeling of incarceration that gradually comes
situation further. The thought of the insurance over one as one enters the hospital. Why incar-
company's waiting until the "midnight hour" to ceration? Because as you enter the environ-
pass along this news was upsetting and fore- ment of the hospital you must gradually
boding. The next day, the insurance company surrender so much, beginning with the beauti-
telephoned and sent a letter to inform me of the ful day, and descend into the mysterious under-
denial of coverage and to inform me of my right ground environment of the building. One might
to appeal their decision. I composed a lengthy arrive first at a subterranean waiting room with
appeal letter with the support of all of my a television as the only link to the outside
cancer doctors, who had given advice on the use world, then be transferred to a smaller waiting
of RFA, and Mr. William McMullen of the room with magazines as the only link. At this
Dana-Farber Cancer Institute. Along with a point one is asked to change into patients'
letter on my behalf from Dr. vanSonnenberg, clothing, and after a brief wait is led down an
we then sent this in to the appeals office of the interior corridor to a room resembling a
insurance company. The appeal was denied. My kitchenette where a large padded chair awaits
wife and I evaluated our options. Since our among various pieces of emergency equipment
family health insurance is purchased through and apparatus. You sit in the chair, have an
her employer, we naively thought that my intravenous device inserted in your arm, and
switching to a different insurance company, one then are escorted to the room that houses the
that would provide coverage, would solve the MRI machine. The machine looks like an enor-
problem. After making a few inquiries we came mous square, shiny, white metal doughnut
to understand three things: standing maybe seven or eight feet high, five
feet wide, and perhaps six feet deep. The dough-
1. Switching, in Massachusetts, could be accom-
nut hole diameter is about the dimension of my
plished only during a specific time period
body measured across from elbow to elbow (I
each year, and we were not in that "window"
am an average-size American male, about 5 ft.
of time.
lOin.), and is located in the middle of the
2. Other insurance company spokesmen either
machine about three feet above the floor. A
did not know if their company covered RFA
padded stretcher resembling a cupped surf
or were not at liberty to say.
board is positioned on a track and centered on
3. It was illegal to change insurance companies
the hole. The stretcher movement is controlled
in Massachusetts, if you were insured through
by electricity, and when activated, can move
your spouse's insurance plan at work.
into and out of the hole in the doughnut with
I was beginning to understand what people you on it in a supine position. I have always
mean when they say there is something wrong gone in feet first on my back. This is when you
with the system. So what happened to create arrive at one of those moments in your life for
the situation that allows me to discuss my which you have had no preparation. This is
ablations? My wife gives some details in her when you lie, flat and straight, in the tube
section. We were informed after losing the (which is the physical presence represented by
appeal at the local level that we still had the hole on the exterior) with tubes attached to
the option of appealing to a higher level in your intravenous device trailing down the tube
Washington, ne. I am happy to say the higher and out the hole, with ear plugs fitted snugly in
level appeal was successful. each ear, with both arms held tightly against
If you want to have an ablation you must first your body, strapped in tightly so as to prevent
have a magnetic resonance imaging (MRI) movement, and find out, if you didn't previously
scan. One is required before the ablation, and know, if you are claustrophobic.
one afterward. This is the main task required of My first time in the MRI machine was with
the patient, and one that I must approach with my eyes open. The inside of the tunnel has tiny
a dauntless and focused attitude. For me, the lamps fitted to the top of the tube in positions
MRI experience is simply an extension of the that seemed to me to be similar to those in an
504 E. vanSonnenberg

airliner. So, if you can imagine yourself as being was painful for me to talk, but not before each
very tiny, it is almost like what you might see of us had agreed to come to the rescue of the
upon waking aboard a night flight to Europe. It other in the event of an emergency by using the
wasn't until I began having subsequent MRIs call button to signal the nurse. He was a
that I began to feel panic about being confined veteran. After the second ablation, I was sent to
even though the total time in the tube for me a cardiac floor after the initial time in the recov-
was only about fifteen minutes. After talking ery area, then sent to the MRI sometime within
about this feeling with Dr. vanSonnenberg, he the next two days after the doctors were satis-
suggested that I close my eyes for the duration fied that I was not having any cardiac issues. My
in the tube, which I have been doing lately and roommate this time was an unpleasant, bel-
this technique combined with counting the ligerent person, about the same age as the first
number of times the magnet is turned on roommate. This one was not, I'm convinced,
(which I have memorized from having done aware of my presence. He kept his TV on all
this about five times) in the process of making night long, woke up shouting during the middle
the image, allows me to make it through the of the night, had some sort of psychotic fit, and
experience with no panic. The technician, who had to be restrained in his bed with straps.
operates the machine, and with whom one must He was hospitalized for a broken leg, which
communicate by way of built-in microphone I thought explained his presence (but this was
and speaker, can make a great difference in the cardiac floor) and the entourage of loud
one's disposition while inside. A few with cheer- obsequious female orderlies. Ostensibly
ful dispositions are very good at this, by reas- present to help him change position and use the
suring the patient and constantly keeping him bed pan, they responded in equal lewdness to
or her informed about what is happening or all of his rancorous remarks and generally
how much is left to do. enjoyed themselves as if they were at a garden
Preparing for the ablation is exactly the party. I pressed frantically on the call button,
same, except one is escorted not to the MRI which was out of order. The hospital, on this
machine but to a CT scan machine. This occasion, was hell.
machine is similar in appearance to the MRI After each of my ablations, and a period of
machine, yet not nearly as large, and the dough- recovery, I have returned to Dana-Farber for a
nut hole is larger, with no appreciable tube. follow-up appointment with Dr. vanSonnenberg
Consequently, when one begins to enter the in the radiology area. Each time he has led me
hole, which is done head first in this machine, deep into the area behind the front desk and into
one quickly exits the other side. The CT scan the room where the radiologists sit before their
machine is used extensively in planning the tra- computer screens and analyze the images trans-
jectory of the RFA probe that punctures the ferred electronically from CT scan machines.
skin and enters the organ and tumor, and in Since the machine is used extensively in locat-
revealing its precise location. The patient is ing the probes that must be inserted in the
anesthetized for the duration of the procedure, tumor, it is possible with computer software,
which can vary in time with the amount of work which links all of the slice-like images taken by
to be done. In my case, this was about three the CT scan machine, to scroll through an image
hours for the first ablation and five-and-a-half of the entire body (only the abdomen in my
hours for the second. case) as if you were looking at a series of cross
Recovery from the procedure is the same as sections. Imagine ordering some sliced roast
it would be for any surgical operation. After my beef at the deli, which is then reassembled as a
first ablation, I was wheeled to the recovery whole as you view each piece. Sitting side by side
area, then to the MRI machine, and then to a with Dr. vanSonnenberg, we reviewed the
room for an overnight stay. My roommate was images of before and after. As he began typing
a man perhaps ten to fifteen years older than I in commands on the keyboard I secretly won-
and talkative. After expressing some initial dered if I would become queasy when looking
curiosity, he gave up on me once I told him it at my own body this way, but when the images
42. Comments from Patients and Their Families 505

finally came up on the Screen my fear gave way into his college dorm to begin his freshman year
to fascination. I would ask, "What is that?" He of college. Along with his mother, the three of
would reply, "That's bone," Or "This is dead us had breakfast at one of those off-campus
tumor here, that's what we want," or "These student-frequented restaurants that serves two
white spots here are your gallstones," or "This dozen variations of bagels with eggs. I remem-
dark area here is air, and this is bowel; here's bered how one of my doctors described the
your stomach." I had been informed by his assis- process of ablating a tumor as a cooking
tant, Dr. Shankar, that lung tumors could also be process, and then it occurred to me that here
ablated and so I asked, "What about the two was a place where one could set definite goals
tumors in my lungs?" to which he replied, "We and make real choices. Here, at school, one
didn't look at those," which I took as an indica- could order the "Big Sur," a toasted bagel with
tion that the operation had lasted long enough a "nuked" (nuked is a word we use at home for
without going to another area. He said ablation referring to something that has been cooked in
worked well for me and that if I lived another the microwave oven) egg sandwiched between
fifty years they could continue doing these over salsa and pepper jack cheese, Or the "Melissa,"
that time span. If the tumors grow back, I was a saucy creation with cooked apples and a
told, we would do this again, and chemother- nuked egg, or a choice of many others. So
apy can be used for anything new. I left Dr. van- maybe, I thought, I could think of the goals for
Sonnenberg's office with a smile on my face, my ablation as "The Big Dave": two liver
feeling like a stalwart member of the radio- tumors, nuked beyond recognition, hold the
frequency ablation fraternity, happy to be alive! bleeding, easy on the pain, served with tender
Nowadays I think of ablation goals at the loving care.
oddest times. Recently, I helped my son move

Patient: BP endocarditis and was hospitalized for two


weeks of antibiotics therapy, and slowly recov-
I am a 77 year young man. In 1984 colon cancer ered at home.
was discovered and the sigmoid section of my On November 15, 2002, I had a thorough
colon was removed. In June 1989 the cancer semiannual checkup that included a CT scan,
had spread to my right lung, and the middle which revealed a 3-cm tumor on my liver. I then
lobe was resected. In 1993 it spread to my left consulted with a hepatologist, who did nothing,
adrenal gland, and that was removed without and asked me to return in three months. This
affecting the kidney. In 1996 the tumor went to made no sense to me. I indicated that I did
my left lung and was successfully resected. not want to wait so long without definitively
Since 1996 the colon cancer has not recurred, knowing what this was. He agreed to see me in
and CEA tests have been normal. I have never two months. At that time, a biopsy indicated that
had chemotherapy, and, until 2003, had not this was a primary liver cancer, not a metastasis
received radiation. from the colon cancer. This doctor recom-
In 2000 I was told that I have aortic stenosis. mended chemoembolization, which I declined.
The valve is nearly closed, but I elected not to I sought a second opinion from Dr. Eric
have surgery. In February 2002 I contracted vanSonnenberg, by which time the tumor had
506 E. vanSonnenberg

doubled in size. He recommended radiofre- of hand. I do not like to think about what
quency ablation, which successfully destroyed may have happened had I not seen Dr.
the tumor without delay. This procedure was vanSonnenberg.
simple, not stressful, and pain free. The doctor's
professional manner, impressive credentials,
Acknowledgments
and concern for me as an individual gave me
great confidence and relieved me from a I would like to thank Mr. and Mrs. Lank for
growing fear that this situation was getting out referring me to Dr. vanSonnenberg.

Patient: Ie Department of Radiology). Chemotherapy


was interrupted for 3 weeks and Iressa
Subject discontinued.
4/02-8/02: Continued with chemotherapy
65 years old, married 43 years, four sons and six (95-FU, Lavamisol and irinotecan).
grandchildren. Owner of computer and sensor 8/23/02: While on vacation, repeat of right
development/manufacturing company. upper quadrant symptoms, and repeat radio-
logic intervention done at Albany Medical
Education Center.
9/13/02: Appendectomy by Dr. Richard
BS Electrical Engineering (1959) MBA (1963). Swanson at Brigham and Women's Hospital.
Pathology report noted cancer cells in the
appendix specimen. Chemotherapy stopped
Cancer History 4 weeks.
12/93: Cancer found on routine physical; left 10/02: Chemotherapy not as effective on lung
hemicolectomy tumor, so changed to 5-FU, oxaliplatin.
1/94-1/95: Outpatient chemotherapy 3/03: Effectiveness of oxaliplatin diminishing
1/00-12/02: CEA rising from 6 to 63 4/03: Dr. Meyerhardt consulted Dr. van-
12/01: Spiral CT showed tumor in left lower Sonnenberg about RFA for the left lower
lobe. Mediastinal biopsy confirmed metastas- lobe lesion in my lung.
tic colon cancer. Also lymph nodes on right 5/29/03: Radiofrequency ablation performed by
and left chest. All pathology from 12/01 and Drs. Sri Shanker and Eric vanSonnenberg
12/93 compared by Massachusetts General at Brigham and Women's Hospital with
Hospital Department of Pathology and thoracic surgeon Dr. Michael Jaklitch in
Brigham and Women's Hospital Department attendance.
of Pathology.
1/02: Chemotherapy at Dana-Farber Cancer
Institute (Drs. Charles Fuchs and Jeffrey
Patient's Comments
Meyerhardt). Also volunteered for Iressa I wanted to be aggressive against the cancer
trial. After 3 to 5 weeks I had pain in my and RFA was an immediate process that, if
right upper quadrant, which resulted in a successful, could really help me. It was also sig-
radiologic intervention to drain the abscess nificantly less invasive than surgery, therefore
from my appendix (Brigham and Women's offering a better recovery potential.
42. Comments from Patients and Their Families 507

I was concerned that the tumor was close to tion I could not change horses in midstream. I
my aorta and perhaps on the aorta. Could they was confident in Dr. vanSonnenberg and in my
get it all? Would I die in the process? own research on the RFA process.
As an electrical engineer, the RFA process As a patient, I think the following improve-
fascinated me. I had never heard of it. I ments could be made:
researched it and could find articles on RFA on
the liver, but none on the lung. One article by 1. If regular surgery is an option, it should be
Gazelle and Goldberg was very helpful as it explained at the earliest opportunity and in
described the tools, salinity effects, process, etc. detail. If surgery is better than RFA, the patient
It made sense to me, but it appeared very needs to know why and how. Also, what are the
precise and tricky to achieve a conversion comparative risks and benefits of each option?
of RF energy to such a precise heat energy 2. The radiologist doing the procedure
targeted to such a small area. should provide the patient with a written
When the procedure was over and I saw the description of the procedure, including the anes-
probes, I was intrigued about their design, the thesia process, the risks, and the tumor location.
materials used, etc. 3. The radiologist should give the patient a
Prior to the procedure, I was confused. When good description of the recovery process. What
talking to Dr. Jaklitch, the thoracic surgeon, should the patient expect? Discharge from
after he saw my CT scans, he said he could "go the hospital one day after the procedure does
in and get the whole thing." I didn't know that not allow for an adequate postoperative
this alternative existed. Without more informa- preparation.

Patient: WBS year later, I learned that the disease had spread
to my liver, and surgical resection was the most
Survival is only one of many competing interests that common therapy for the disease; however, it
influence thought and action. was a procedure from which recovery would be
difficult for a person of my age-I was 80 years
William James old.
My vocations were in the fields of engineer-
ing and architecture, and I was, and still am,
"Ten percent of liver metastases patients have accustomed to gathering information and
resectable disease, and surgical resection has making decisions for myself. However, there is
yielded a 20% to 40% five-year survival rate." one exception to this rule: when it comes to
This statement appeared on one of the pages medical decisions, I usually follow the recom-
yielded by the National Cancer Institute's Web mendations of my doctors. This time I did not;
site in March 1999 when I typed the following with a 20% to 40% 5-year survival rate, the
search words: resection of hepatic metastatic odds were too great. So I went back to the Web.
colon cancer. Survival was uppermost in my mind, and
In June 1998, 12 inches of my ascending colon soon I had a three-ring binder overflowing with
were removed surgically because a moderately Web-based reports on the results of chemo and
differentiated colonic adenocarcinoma had percutaneous ablation studies, articles from
invaded the pericolonic fat in my colon. One newspapers and magazines, and notes on sug-
508 E. vanSonnenberg

gestions from friends. After reading all of the I signed it knowing I had made the right
material, I concluded that things did not look decision.
good for me; cancer cells were in my blood-
stream and taking root in new places. It was
time I gave thought to avenues other than those
Not for the Claustrophobic
that would just lead to my survival. My participation in the study began with some
Clinical studies offered such an avenue. Here misgivings. The first was the location of the
I would be able to contribute to science, and, if treatment facility. One reaches the areas in
the treatment worked, so much the better. I BWH where the CT and MR machines are
went back to the computer and there I found a installed by taking a down elevator to below-
study that caught my eye. It was at Brigham and grade floors and walking a long hall with a low
Women's Hospital (BWH) in Boston; its ceiling, the occasional fire door, and walls hung
purpose was to evaluate the effectiveness of with abstract prints. While it is well-lighted, the
using a needle-like probe to apply cold cell light is not the kind of light I like; I am a
killing temperatures to a tumor. It was called natural-light person.
"MR-guided cryoablation of liver tumors." The The reception and imaging areas exhibited
bottom line-I would be a guinea pig for tools the same characteristics as the hall, but they
that make iceballs of body parts. received additional light from the cheerful and
accommodating people that worked there. The
machines themselves are another story-they
I Go for It can be intimidating. Spelunkers and cave
I contacted the principal investigator for the dwellers will be right at home in an MR
study and, soon thereafter, my wife and I drove machine. I told myself that I had to compare
the 170 miles to the hospital, where I under- this with an operating room, and, thankfully,
went physical exams and imaging studies and the machines won hands down.
completed reams of paperwork. Parking and Actually, I had experienced CT and MR
lodging were close by and their costs were machines on more than one occasion before I
reasonable. The personnel were friendly and went to BWH, but this time someone was going
informative. to poke at me while I was lying in them. As to
The findings of the exams and imaging their being in the basement, this seems to be
studies were that I had a medical history of where imaging machines are installed in
kidney stones, polyneuropathy, monoclonal hospitals. Being a pragmatist, I began to warm
gammopathy of unknown significance, and up to the environment.
myelodysplasia, and that I presently had one
hepatic metastatic colon cancer tumor that
My Tumor Is Frosted
measured 4 x 4.5 cm. All of these diseases
notwithstanding, I was eligible for the study. Three days after my 81st birthday, with a light
My first surprise was learning that radiolo- heart, I took the elevator down, walked the
gists would perform the procedure. I thought hall, donned the gown, boarded a gurney, and
radiologists just operated imaging machines greeted the anesthesiologist. After he explained
and read the pictures they produced. I soon the procedure he would follow, the radiologists
learned that it was this expertise that made it came in. They described what they were going
possible to perform the type of cryoablation to do, and asked if I had any questions. I said I
procedure that I was to receive. was ready and a sedative was administered.
I was given a form that thoroughly described I kissed my wife, was taken to the MR machine,
the purpose of the research, the study proce- and soon thereafter was in la-Ia land.
dures, and possible risks and discomforts. There The next thing I knew, I was lying on a gurney
was a place for me to indicate my willingness to and wondering who had inserted the catheter
participate in the study and place my signature. for draining my urine. My wife was at my side,
42. Comments from Patients and Their Families 509

and for the next few hours I dozed, ate some benefits, and alternatives were discussed with
yogurt, and sensed the coming on of slight dis- me; the fact that it was experimental therapy
comfort in my belly. I was moved to a hospital was noted, and I gave my written consent to
room where I spent the night trying to get com- going ahead. The procedure followed the same
fortable-I wish someone would invent a dif- lines as the first. Again, I recovered with no sig-
ferent way to collect patients' urine when they nificant aftereffects. An MR scan 4 months
are asleep. At 4 a.m., I was taken to the base- later showed a 4.1 x 3.4cm crescent-shaped
ment and put in the MR machine. Both the area that might be residual or recurrent neo-
technician and I were grumpy, and the whole plasm. A biopsy performed a month later read
thing was a traumatic experience. positive for malignancy.
But I lived, and later in the morning, the radi- Seven months after the first treatment, the
ologists came to my room and told me that they team concluded that my liver function was
were happy with the results of the cryo proce- normal; the active portion of the tumor was
dure. For several reasons, they had stopped smaller; an increasing CEA might indicate
after treating 80% to 90% of the tumor: I had increasing tumor activity in other areas; the
been "under" for 5 hours; the untreated portion tumor was still resectable; and more cryother-
of the tumor was so close to the diaphragm that apy in the vicinity of the diaphragm would be
freezing it was risky; and, finally, they wanted to dangerous. It was suggested that, if I wanted
have some MR scans done to see whether the further percutaneous treatments, they would
procedure was working. I no longer felt like a like to give RFA a try. There were two draw-
guinea pig; these men really wanted to kill my backs: I would not be in a clinical study, and
tumor. because the tumor was close to the diaphragm
Probably because I have myelodysplasia, the RFA treatment might just be a way to keep
my red blood cell count dropped to the high the tumor at bay.
20s after the procedure. The next day it was in There were other persons in the cryo study
the low 30s (where it had been when I was and more to come, so the investigators could
admitted). I was discharged with the same not tell me whether they had achieved their
discomfort in my belly, a lack of energy, and goal. However, I left the study believing that all
Band-Aids where the probes had entered my of the tumor would have been killed if it had
abdomen. not been so close to the diaphragm. As to
We drove home, and this time I occupied the whether I had achieved my purpose in becom-
passenger seat of our car. I slept soundly that ing a participant in the study, I believed I had
night, and the next day I rested while listening contributed to science and had received a
to a books-on-tape version of a Civil War story bonus in the form of a reduction in the size of
titled The Killer Angels. I could not help reflect- my tumor. The latter benefit gave me some time
ing on the quality of medical treatment during to pursue a clinical study of a different nature.
the 1860s; I went so far as to compare it with
the surgical removal of a metastatic tumor
versus killing the cells with a probe.
My Experiences with
Radiofrequency Ablation
My choices for a new clinical study appeared
Following Up to me to be limited to ones that were chemo
The next week, I felt great and went back to oriented. Believing that chemo would be time-
Boston for a checkup. The MR machine showed consuming and debilitating, especially so with
that healing was progressing, and within the my myelodysplasia problem, I began looking
portion that had been treated was an area that for a study that was close to home where I
may not have been frozen; it measured 1 x 2 em. could, more comfortably, put up with the effects
Two months later, I went back for another of chemo. I found none, and took up BWH's
cryo treatment. The procedure and its risks, offer of RF therapy.
510 E. vanSonnenberg

My Palette by was night and the next day trying to satisfy the
hospital's fluid discharge requirements, and,
If one has a choice in the radio having done so, went home the third day.
frequency used in my RF ablation Two weeks later, a CT scan showed a 6 x
treatment, I would like one that 4 cm abnormal area with a 1.3 x 1.3 cm area that
plays classical music. the radiologist believed was residual tumor. The
radiologist noted that, at best, these dimensions

~ o
were "guesstimates" because so much had
happened to that area of my liver, it was diffi-
cult to distinguish between what might still be
alive and what was dead.
Three months after the RF treatment, a CT
scan showed the live portion of the tumor was
bilobed; one lobe (near the vena cava) mea-
sured 2.7 x 2.4cm, and the second lobe (near
the diaphragm) measured 2.9 x 2.6cm. The
tumor was being held at bay.
The radiologist said that with another treat-
ment, they might kill all of the cells in the two
lobes except those in a small portion near the
FIGURE 42.1. An engineer patient's KF preference. diaphragm; so 5 months after the first RF treat-
ment, and 13 months after I received my first
percutaneous treatment, I went back for what
would prove to be my last experience with
percutaneous ablation.
I gathered enough information on RFA to It was a nice warm day in June 2000 and all
know how it worked and what would be done went well with the RF procedure. However, this
once I climbed on the table of the CT machine. time my recovery was slower; my hemotocrit
I learned that RF therapy sometimes is done on count had dropped to the middle 20s right after
an outpatient basis with local anesthesia. This I came home, and would not rise again. This
was not for me; I wanted general anesthesia and condition may not have been caused by the RF
a short hospital stay. The most optimistic report procedure-low hemotocrit counts are also a
I found on RFA said it yields 65% to 75% effi- product of myelodysplasia.
cacy in the control of tumor growth in metas-
tases. Those were odds I could live with.
Ten months after I had entered the cryo
Since Then
study, I returned to Boston where the anesthe- In July my oncologist started me on weekly
siologist and radiologist explained what they shots of Procrit in an effort to bring my blood
were going to do, the risks, and so forth. I asked count back to the middle to low thirties. Occa-
some questions (Fig. 42.1). I found the CT sionally, the count would exceed 36 and I would
machine to be less intimidating than the MR, stop for a while, but only for a short while-
and was relaxed knowing the radiologist was myelodysplasia had begun to take its toll. On
not exploring a new field of science. I signed top of that, a PET scan in September showed
the usual release forms, and the treatment was an "intense tracer uptake in a large area of the
administered. right hepatic lobe and in the anterior aspect of
The CT machine saw a 3.5 x 4.2cm lesion on the right lower chest wall plus a small area of
the right lobe of the liver near the dome of the uptake in the posterior aspect of the right hilum
diaphragm. The radiologists performed two 12- of the lung." The lower right chest wall is adja-
minute RF applications. I recovered from the cent to the points of entry on my abdomen for
anesthesia with no complications, spent that the cryo needles. The consent form I had signed
42. Comments from Patients and Their Families 511

for admittance to the cryo study noted that that influenced my thoughts and actions. I tried
spreading tumor cells along the path of the chemo by injection (it all but knocked my socks
needle is exceedingly rare. My mother believed off); treated bouts of erratic heart beat and
I was one-in-a-million, so it is befitting that my shingles; battled two squamous cell eruptions
tumor cells would hitch a ride on the cryo on my head; lost much of the use of my left
needle. hand and arm from reflex sympathetic dystro-
While time seemed to be getting short, I had phy; strapped on a truss to hold two hernias in
not lost sight of my desire to participate in check; underwent surgery and radiation in an
another clinical study. I found one that I effort to remove the lesion on my chest wall;
believed I could weather and, just before punched another hole in my belt to help with
Thanksgiving, I went to Memorial Sloan- the problems that come with a 25-pound weight
Kettering Cancer Center to talk about hepatic loss; and grimaced many times over while
arterial infusion of chemo substances using an Procrit was being administered and blood
implanted pump. My hopes were dashed when was being drawn for CBCs.
the administrator said the cryo and RF treat- All the while, rogue cells in my body keep
ments made me ineligible. doing their thing and reminding me that maybe
It is now October 2002. During the past two now is the time to let nature take her course.
years, survival became the dominant interest On the other hand, maybe it isn't.

Patient: MH pain and why its presence is so difficult. It is a


way that the body communicates, and cannot
Fiance: PW just be masked or ignored. It needs to be heard
and addressed for any healing to occur.
My fiance passed away on March 20, 2002, of Although this was an extraordinarily difficult
a chondrosarcoma that had finally found its lesson, it forced us both to look very closely at
way to his lungs. My story is not your traditional our priorities and make progressive choices
success story in that he did not survive, but for improving our quality of life. It is not
our travels in the realm of cancer taught us enough to simply fight for more days. The true
both some invaluable lessons about medicine, fight is in how to enrich those days.
accessibility of treatment options, the health He had been in remission from a non-
care industry, the brutal reality of insurance Hodgkin's lymphoma in the stomach for fifteen
arguments, and most importantly, ourselves. years when he was diagnosed with a chon-
We traveled through a world of healing drosarcoma in the left shoulder approximately
together, and although he did not survive one year before we met the team at Dana-
the illness, he healed unlike any other. Our Farber Cancer Institute. The first year after he
journey through this world of cancer has was diagnosed was a year of surgeries, appoint-
taught me more than I possibly could have ments, sickness, chemotherapy, nutrition coun-
imagined. seling, more sickness, more chemotherapy,
We learned that in order to heal, we must hospital stays, new doctors, rehabilitation, stem
love every part of the sickness, or gift, as we cell transplants, more chemotherapy, more sick-
later referred to it, including the very real and ness, and more chemotherapy. We were told
very loud aspect of pain. We traveled down from the beginning that sarcomas rarely react to
many roads to try and understand the idea of chemotherapy, and yet it was the treatment of
512 E. vanSonnenberg

choice time and time again. The side effects of never receiving relief. He was constantly fight-
these treatments consumed him, and he grew to ing the drowsiness, depression, constipation,
fear his own mind during the time he was subject and delirium that the drugs were offering.
to these sessions. After a year and a half of this Every day we were increasing the dosages of
treatment, he was left with the original tumor in methadone, fentanyl, oxycodone, Neurontin,
his shoulder, a new tumor in his pelvis, several and Dilaudid. Our physicians were simply
tumors in his lungs, intense pain in his pelvic handing over stronger and stronger prescrip-
area, and absolutely no immune system. tions. Despite our faith, we felt as though we
Still, he was only 24 years old and had were spinning out of control, the illness was
a huge will to live. I was committed to doing starting to give the impression that it was taking
everything that I could to support him. over. The only other options we were given
Throughout our many hospital visits I was were more chemotherapy or the wildly attrac-
always surprised at the physicians' need to tive radiation therapy.
remind us of "the reality of the situation," or to From earlier discussions about the resistance
be more frank, that he should be prepared a sarcoma has for radiation, we knew it was
to die. This was the priority, rather than the being suggested as a last resort. We were told
discussion of treatment plans. It felt as though that it would not save his life but perhaps
our hope was perceived as denial. prolong it. It would prolong his life, but, we
In truth, our reality was far different from suddenly realized, what life? Is it life when you
that of the physicians. Although we were com- cannot even get out of bed or go to the bath-
pletely aware of the possibility of death, he was room without doubling over in agony? Is it life
still alive and should be treated as such. We when every position is uncomfortable and
could not understand the change in attitude nothing smells or tastes appealing? Is it life
after he was bestowed the title of terminal-a when you barely have energy to walk to the
label that haunted us every day. In our world, next room, let alone spend time with your
that label meant we fight harder and use our friends? Is it life when you are filled with crazy
time to the fullest. In the world we suddenly dreams from the drugs that you are forced to
found ourselves, it meant not only slowing take every four hours, when the only vacation
down but giving up. It was as if the entire you have to look forward to is your next trip to
Western medicine industry shut down. Their the radiation room? He deserved a night of
efforts were exhausted, and they wanted no sleep, and I deserved a day free of his winces,
part of any new, alternative, or unconventional pill popping, and screams of frustration.
ideas, regardless of their apparent success. The I stepped up my extensive research regard-
insurance companies ignored our pleas for help ing pain management and must have explored
in our desire to try anything other than the two every search engine available. Finally, I came
mainstream and obviously ineffective choices across an article that was written about a
of treatment-chemotherapy and radiation. physician, Dr. Sridhar Shankar, at Dana-Farber
This did not sit well with us at all. I watched him Cancer Institute, regarding the fairly new world
wake up every morning to the immense emo- of cryotherapy and radiofrequency ablation.
tional pain, caused in part by the fear of dying, We had enough of simply prolonging the
accompanied by the infamous physical pain. We illness, so I composed a letter that would soon
were never told about radiofrequency ablation lead us into the world of radiofrequency abla-
as a pain management source, let alone a tion and introduce us to the amazing team at
possible treatment method. This we were Dana-Farber Cancer Institute. The letter read
fortunate enough to discover on our own. We as follows:
began our own search, with a true physical yelp
leading the way. I made it my sole mission, as Dear Dr. Shankar,
any teammate would do in my situation, to get First let me thank you for the phone conversation
him out of pain. we had last Friday afternoon and for your patient
We had already spent a year and a half voice regarding my seemingly desperate situation.
chasing it with a wide variety of narcotics and I am sure that you and your facility get many similar
42. Comments from Patients and Their Families 513

calls from stressed patients and you must know that At this time we decided to leave Sloan-Kettering
your attention and knowledge is greatly appreciated. and seek out a sarcoma specialist. My fiance began
I have included a general patient history of my treatments under the care of Dr. Gerald Rosen of
boyfriend's case, along with relevant images that I St. Vincent's Cancer Center. Immediately he began
feel will be helpful to you. I will need these returned more chemotherapy treatments (methotrexate) with
to us either by mail or in person, depending on the some success. He was given four rounds (I-day injec-
course of action that is decided on. tion and I-week recovery) of methotrexate, and then
Forgive me if I seem long winded, but at this time a high dose of carboplatin (4-day injection with a 2-
I feel the need to just fill you in on the basic events week recovery), including stem cell transplant. This
of the past year to give you perspective as to why we ended about one month ago, and a follow-up PET
are looking to you for help. scan showed no response.
Last fall my boyfriend was feeling pain in his left The next plan scheduled for the new year is radi-
shoulder region and went to his primary care physi- ation therapy, with surgery still hanging in there as
cian who diagnosed a pulled muscle, and sent him the goal, but now in the pelvis and the chest. This
home with a cortisone shot. The pain persisted and does not sit well with us, and we fear that time is
brought him to the hospital. He was given an x-ray crucial.
and it was determined that there was a mass in his So, with two major surgeries behind us, multiple
shoulder that was suspected of disease. He had prior rounds of intense chemotherapy still haunting
non-Hodgkin's lymphoma as an eight-year-old, and us, fear of paralysis or limb salvage, the pressure
was in remission for 15 years. His primary physician for radiation, and a 24-year-old still waking up in
then referred him to Memorial Sloan-Kettering pain every day, we are looking to cryotherapy/
Cancer Center, where he began consultations and thermotherapy to provide any sort of push in the
had a biopsy of his chest on December 27, 2000. The right direction. As you can tell, time is valuable to us
diagnosis was, shockingly, chondrosarcoma of the and please be aware of how incredibly grateful we
chest, and the plan was surgery in the beginning of are that you have even taken the time to read this
the new year. This never happened. letter and offer any advice you can.
He experienced shortness of breath, which took Over the phone you asked me to give my expec-
him to Memorial Sloan-Kettering ER on December tations as to what I thought you could do. I would
31,2000. He stayed at the hospital for approximately not be true to myself or my heart if I did not tell you
one month as an inpatient. They put him on heparin I expect tumor shrinkage, cease in pain, and a future
to treat what was thought to be a blood clot in his of remission. However, that being said, I honestly
heart, and an immediate open-heart surgery was per- believe that everything happens for a reason, and
formed in early January 2001. This surgery surpris- that finding your facility and your research is simply
ingly showed tumor in the heart (NOT a blood clot) just another step in our path as we continue to expe-
that was successfully removed. rience the world of cancer.
Approximately one week after open-heart I thank you again for you attention to our
surgery, a PET scan and MRI revealed tumor around case, and please call us as soon as possible with any
the spine, with a paralysis scare that took him into questions, concerns, advice, and/or recommenda-
surgery. Tumor was removed from around his TI. tions. I will give you a follow-up call to make sure
This was also successful, and he began the recovery the films were received. I wish you success in your
process from both surgeries. endeavors.
About a week into the recovery process,
chemotherapy was started to treat the tumor still in
his chest region with the original long-term goal of
surgery still present. He underwent chemotherapy
(Adriamycin and ifosfamide) on and off for about Respectfully yours,
4 weeks. A follow-up CT scan did reveal some tumor PW
response. The same course of action was continued.
After another 4 to 6 weeks, they did another CT and
This letter was well-received and for the first
PET scan, and the chest tumor had not only ceased
response to the chemotherapy, but a new tumor was
time we felt like we were not fighting this alone.
discovered (approx. 6cm) in the pelvis region. This We immediately booked two train tickets to
was never mentioned before, even though there was Boston and along with his pillows to sit on, as
complaint of pain in the lower back. The choice of many of our imaging studies and medical
treatment was the same chemotherapy that had reports that we could carry, and our incredible
ceased to work on the chest. optimism, we were off to the Dana-Farber
514 E. vanSonnenberg

Cancer Institute to learn more about radiofre- and harder, as the illness was getting larger and
quency ablation. more severe. The need for pain manage-
We arrived and were instantly impressed ment was crucial for the quality of his moments,
with the professional greeting we received, an and we began counting the days until the
attitude that truly never ceased throughout our ablation.
entire time there. We met with Dr. Eric van- The morning of the ablation was full of antic-
Sonnenberg and began our first step to pain ipation, excitement, and fear. He and I, along
control. Dr. vanSonnenberg and Dr. Shankar with his family, were briefed again on the
took the time to sit with us, so that we could sequence of events, and then the prep for anes-
discuss our options. As we gained knowledge thesia. I helped him as he climbed onto the
about the basic events surrounding the proce- gurney and was pushed into the CT scanner.
dure, we also gained hope for a future: a crucial The procedure lasted approximately four
and often overlooked tool in pain management. hours, involved two tiny incisions about an inch
We were amazed at the possibilities that abla- wide, and six different ablations, each of them
tion offered. As we discussed, it would actually the size of a large apple. Immediately after
heat the tumor from the inside and essentially the procedure I met him in the recovery
dehydrate the cancer cells. The cells would die, room and although still very stiff, he was fine.
and the body would rid them in the natural He was very pleasant and able to speak to me
fashion. This in turn would cause the tumor to about his experience. We were taken to a
shrink and, it was hoped, help with the pain. hospital room where we stayed for two nights
This was a treatment that entailed no vomiting, of healing and monitoring vitals. The recovery
lack of appetite, prolonged side effects, or process from the ablation was nothing com-
amputation (as surgery would have called for). pared to our prior experience, and he was
It would involve a few days in the hospital, as walking around the next morning and had a
the cancer was very involved, and a Band-Aid huge appetite. He was given a follow-up MRI
on his hip. We found ourselves suddenly filled that showed a great deal of dead tumor that
with enthusiasm as we plotted a game plan we would take some time to pass through. He was
could believe in. still in a great deal of pain, but the pain itself
The tumor that we focused on, as it was was significantly different.
causing the most pain, was located in his pelvis. By the time we found our friends at Dana-
It involved his pelvic bone and was creating a Farber, were fit into the mainstream schedule of
great deal of pressure on his testicles. He found the hospital, and fought with the insurance
it impossible to sit for any period of time and companies to help us receive treatment, he had
carried a pillow everywhere. The tumor was multiple tumors and his disease was incredibly
also pushing on the sciatic nerve, which sent advanced. The ablation was successful in a
shooting pains down his entire leg. He had a variety of ways. He no longer had pain down his
very high tolerance for pain and lived with a leg and he was able to sit up and walk around
constant level-4 pain (on a scale of 1 to 10) with less difficulty. The tumor also significantly
throughout his experience with sarcoma. At this decreased in size for the first time since the
time, however, he was fighting with a pain that diagnosis. Unfortunately, it was growing at such
was rising to levels above 7 and 8, despite the a rapid pace that the remaining part inside his
constant increase in medications. He was trying bone was still causing pressure. The ablation,
to cope with the everyday struggles, while he however, allowed him to maintain his state of
became weaker and weaker in mind and body. mind, which was the most important aspect to
Physical pain and the emotional stress that him, and he was given a sense of hope that
joins with the fears of having a terminal illness would carry him through his days. He experi-
create a devastating perception of the self and enced apparent relief, and he was able to laugh
the world around you. With each passing again. He had energy to continue his alterna-
day and with each cry for help, he was trying to tive medicines, visit with friends and family, and
trust each moment. This was getting harder make conscious choices regarding his next
42. Comments from Patients and Their Families 515

course of action. We had about two weeks after everyone who enters a hospital needs an advo-
that first ablation and were actually planning cate. We discovered that you also need a great
another on the remaining part of the tumor deal of money to receive treatments deemed
when the lungs had other plans. The tumors in "unconventional" that could advance your
his lungs were causing him to experience short- prognosis and help keep you out of pain.
ness of breath and we found ourselves travel- Inspired by his courage and faith, by his sheer
ing down another path. I wish that I could say determination to lead an uplifting and fulfilling
that this procedure saved his life, but instead, I life, I agreed to share our story. I want every-
can only wish that this information leads to one to know what is available to them-all of
other stories of that sort. It was not that the the options, treatments, and possibilities, not
ablation did not work. Unfortunately, we just just those currently being advocated by main-
found our miracle a little too late. stream physicians and insurance companies. My
We live in a world where we are taught to wish is that anyone in a similar situation (and I
give our complete trust to those who know am aware that this includes millions of people
"more"; to give away our own power simply every day) could access any and all information
because the efforts of the "experts" have been that will help them to honor, in sickness and in
exhausted. We discovered, the hard way, that health, every moment of their lives.
Index

A Adenocarcinoma American Society of


Abdominal viscera, subscapular mammary, cryotherapy for, Anesthesiology, 66-67, 70,
tumors adjacent to, 210 436 74
Abdominal wall, radiofrequency of prostate gland, 151 Anastomoses
ablation-related burns to, Adenoma, pleomorphic biloenteric, 441, 442-443
476 laser-induced interstitial utero-ovarian, in uterine
Ablation generators, tissue- and thermotherapy for, artery embolization,
organ-specific, 287 239-240 416-417
Ablation therapy. See Tumor parapharyngeal,240-241,245 Anesthesia, in tumor ablation
ablation therapy Adenomyosis, as uterine therapy, 64-75
Abscess leiomyoma mimic, 413 continuous quality
of bone, 393-394 Adolescents, osteoid osteoma in, improvement in, 73-74
cr-guided drainage of, 116 389-390 equipment for, 60-73
hepatic Adrenal gland, intra-ablation hypotensive, 301-302
cryotherapy-related, heating of, 443 MRI-compatible delivery
447-448 Adrenal lesions, radiofrequency systems for, 154
ethanol injection ablation of, 445-446 in osteoid osteoma patients,
therapy-related, 441 Adrenocortical carcinoma, 287 395-396
microwave coagulation Adriamycin. See Doxorubicin in pediatric patients, 498, 481
therapy-related, 451, 475 Agency for Health Care Policy procedure room design for,
radiofrequency and Research (AHCPR), 70-73
ablation-related, 442, 476, 355 quality assurance in, 73-74
485,499 Airway examination, by reimbursement for, 74-75
prostate cryotherapy-related, anesthesiologists, 66 remote-site administration of,
449 Albany Medical Center, 514 72-73
pulmonary, radiofrequency Alcohol selection of techniques in,
ablation-related, 117 hypoattenuating activity of, 67-68
Abstracts, example of, 17 94 standards for, 65-67
Acetic acid injection ablation iodized, as contact medium, for monitoring during
therapy, 10, 24-25 196 procedures, 66-67
Acoustic shadowing, in Alcohol injections. See Ethanol for postanesthesia care units,
cryotherapy, 100 injection therapy 67
Acoustic streaming, 296 Alcohol intoxication, ethanol for postoperative visits, 67
Acute respiratory distress injection therapy-related, for preoperative visits, 65-66
syndrome 198,441-442 types of, 68-70
cryotherapy-related, 446 Alveolar condensation, 113 conscious sedation, 68-69
radiofrequency American Joint Committee on general anesthesia, 67, 69,
ablation-related, 443-444 Cancer, 341, 342 357

517
518 Index

Anesthesia, in tumor ablation Arteriovenous malformations, radiofrequency


therapy (cant.) 369 ablation-related
local anesthesia, 67, 68 Arthritis, osteoid stenosis/strictures of, 443,
monitored anesthesia care osteoma-related, 394 485
(MAC), 69 Artifacts, in interventional MRI, thermal damage to, 442
regional anesthesia, 69 153 Bile leaks, hepatic
Anesthesiologists, involvement in Ascites, microwave coagulation radiofrequency
tumor ablation therapy therapy-related, 224 ablation-related,485
advantages of, 54-65 Aspiration, of gastric contents, Biliary tract, dilation of, 115, 117
disadvantages of, 65 356 Biliary tract, radiofrequency
Angiogenesis, in tumor growth, Aspirin, as osteoid ablation-related injury to,
41-55 osteoma-related pain 210
historical background of, 41-42 treatment, 390 Bioheat equation, 27, 30, 206,
inhibitors of, 41-42, 44, 45 Azwell microwave ablation 208,286-287
as cancer treatment, 45-47, system, 88 Biomarkers, 184
167,209,298 Biopsy
direct vs. indirect, 42-47 B differentiated from tumor
endogenous, 47 Balloons, as protective adjunctive ablation therapy, 108-109
hyperthermia-enhancing technique, in tumor fine-needle
effects of, 50 ablation, 373 comparison with tumor
inhibitor/stimulator '"switch" Balloon vascular occlusion, in ablation, 347
in, 44-45 radiofrequency ablation, complications of, 440
in metastases, 44-45 287-288 of lung cancer, 355
steps in, 42-43 as bile duct injury cause, 301 prior to renal cell carcinoma
stimulators of, 41-42, 43, 44 of the hepatic artery, 6, 32-33, ablation, 345-346
Angiostatin, 45, 47, 49 209,301 of renal cell carcinoma, 347,
Angiozyme, 48 limitations to, 33 348
Animal models, of MRI-guided of the portal vein, 442 of gastrointestinal stromal
radiofrequency ablation, tumor modeling and, 287 tumors, 150
173-174,176 Basic fibroblast growth factor of hepatocellular carcinoma,
Antiangiogenesis therapy, 45-47, (bFGF),41-42,43,45,50 327
177,209 Beckwith-Wiedemann syndrome, mediastinal, 107
nanoparticle-based, 298 hepatoblastoma MRI-guided,149-151
Antibiotic prophylaxis, in tumor associated with, 501, 502 pelvic, 107,371
ablation therapy patients, Benzodiazepines prostate, MRI-guided, 149, 151
440,441 use as conscious sedation, 68 retroperitoneal, 107
in pediatric patients, 499, 501 use as monitored anesthesia single-trajectory, 184-185
Anticoagulants, as care (MAC), 69 ultrasound-guided
radiofrequency ablation Berchtold radiofrequency freehand technique in, 99
contraindication, 357 ablation system, 81-82 hepatic, 98-99
Antithrombin III, antiangiogenic Berchtold radiofrequency laparoscopic, 99
activity of, 47 needles, 7 Bladder metastases,
Argon gas-based cryotherapy Bevacizumab, 48 radiofrequency ablation
systems, 83-84. 252, Bile ducts of,213
253-254,484 balloon occlusion-related Blankets, hyopthermic, 499, 502
Joule-Thompson effect in, 83 injury to, 301 Bleomycin
Argon/helium gas-based cryotherapy-related injury to, in combination with
cryotherapy systems, 253,447 electroporation, 295
84-86 laser-induced interstitial cryotherapy-enhanced uptake
Endocare system, 84-86 thermotherapy-related of,294
Galil system, 84, 85 injury to, 450 Blood flow, during thermal
Arsenic trioxide, 33 microwave coagulation ablation therapy, 8
Arteriography, pelvic, 414, 415, therapy-related strictures effect on ablation therapy
416 of,224 efficacy, 101
Index 519

effect on ablation volume, 183 Brachytherapy C


management of. See also for lung cancer, 361 Cancer. See also specific types of
Balloon vascular occlusion for prostate cancer, 161-162 cancer
with pharmacologic Brain metastases, radiofrequency angiogenesis in. See
modulation, 33 ablation of, 209, 213 Angiogenesis, in tumor
with Pringle maneuver, 6, Brain tumors, cryotherapy for, growth
32-33,101-102,209,212, 154 antiangiogenic therapy for,
213 Breast cancer/tumors 45-47,167,209
Body wall masses, CT-guided cryotherapy for, 154,422-427, nonparticle-based, 298
cryotherapy for, 257 428,436-438 symptoms of, 21
Bone, abscesses of, 393-394 as cryoprobe-assisted Canstatin,47
Bone metastases lumpectomy, 425 Capillary blood vessels, in tumor
breast cancer-related, 312 of fibroadenomas, 422-425 growth. See Angiogenesis
external radiation therapy for, as primary therapy, Carbon dioxide, as protective
355 425-426 adjunctive technique, 373
pain management in. See Bone ductal carcinoma in situ, 426, Carbonization, of tissue
pain management 429 in laser-induced interstitial
radiofrequency ablation of, 213 focused high-intensity thermotherapy, 235
complications of, 445 ultrasound therapy for, in radiofrequency ablation,
Bone pain management, 377-388 160-161,428,440-436 159,169,205,207
CT use in, 110-111 imaging of, 184 prevention of, 170
with ethanol injection therapy, laser-induced interstitial Carcinoembryonic antigen
378 therapy ablation of, 159, (CEA), 483, 486
with external-beam radiation, 440 Carcinoid syndrome, 333, 334,
377 metastatic to 338,469
with laser-induced interstitial bone, 312, 371, 377 Carcinoid tumors
thermotherapy, 378 the liver, 98, 212, 303, 318 appendiceal, 469
with radiofrequency ablation, the lungs, 312, 354 metastatic, 332-333, 338, 468
378-386,463,464 soft tissue, 371 small-bowel, 332
Bone tumors MRI-guided biopsy of, 151 Carcinomatosis, peritoneal, 469
CT-guided ablation of, mucinous, 429 Cardiovascular disease patients,
110-111 postablation CT imaging of, anesthesia in, 65-66
MRI-guided cryotherapy for, 115 Catheterization
156 radiofrequency ablation of, of chest, in lung cancer
postablation CT imaging of, 168,428-440 patients, 357
117 with chemotherapy, 303 urethral warming, 373
postablation MR imaging of, complications of, 446 Cavitation nuclei, 296
117 equipment for, 430 Celecoxib,47
Bonopty penetration set, 396, 397 in liver metastases, 212, Celiac plexus nerve blocks, 67,
Boston Scientific radiofrequency 303 69
ablation system, 79-80 ultrasound guidance of, Cell death. See also Coagulation
Bovie electrocautery device, 429-438,439 necrosis; Necrosis
262 surgical management of, 428 cryotherapy-related, 252-253,
Bovie knife, 3-5 tumor margin localization in, 261
Bowel. See also Colon; Small 183 imaging confirmation of, 112
bowel tumors Brigham and Women's Hospital, Cell surface antibody therapy, for
cryotherapy-related injury to, 510,514,489,491 hepatocellular carcinoma,
253 Burns 484
Bowel metastases, microwave coagulation Celon radiofrequency ablation
radiofrequency ablation therapy-related, 451 system, 82-83
of, 213 MRI-related, 172 Chemical ablation therapy, 23,
Brachial plexus injuries, in radiofrequency 24. See also Acetic acid
anesthetized patients, ablation-related, 110, 443, injection ablation therapy;
73-74 444,446,476,489 Ethanol injection therapy
520 Index

Chemoembolization via hepatic arterial infusion, Circulating endothelial


as hepatocellular carcinoma 467-468 precursors (CEPs), in
treatment, 6, 218, 243, 322, for metastatic neuroendocrine angiogenesis, 42, 43-44
323,326-327,468,473,484 tumors, 468-469 Cirrhosis
in children, 501 for pancreatic cancer, 469-470 as contraindication to
with ethanol injection for pediatric hepatocellular ethanol injection therapy,
therapy, 266-267 carcinoma, 501 198
with laser-induced percutaneous intratumoral, liver resection, 218
interstitial thermotherapy, history of, 9-10 hepatitis B-induced, 209, 211
269 with radiofrequency ablation, hepatocellular carcinoma
with microwave coagulation 287,303-304 associated with, 472,
therapy, 268-269 liposomal, 35-36 483-484
preoperative, 267, 268 synergism with ethanol injection therapy
as liver metastases treatment hyperthermia in, 35-36, for, 24, 200
comparison with 304 radiofrequency ablation of,
laser-induced interstitial via implantable pump, in liver 209,211
thermotherapy, 243-244 resection patients, 59 ultrasound imaging of,
following surgical resection, Chest tubes, CT-guided 141-142
269-270 placement of, 111 ultrasound screening of, 201
with radiofrequency Chest wall, lung cancer invasion liver metastases associated
ablation, 303, 304 of, 354-355 with, 135
in liver transplantation pain control in, 371 liver tumors associated with,
recipients, 468 Children 441
as neuroendocrine hepatic chemoembolization therapy in, Cisplatin, 269, 484
metastases treatment, 501 Clear cell carcinoma, renal, 342
334 chemotherapy in, 501 Clonazepam, 509
with radiofrequency ablation, osteoid osteoma in, 389, 390, Clopidogrel, 463
209,287-288,303,304 498-481 Coagulation necrosis, thermal
transcatheter arterial (TACE) radiofrequency ablation ablation therapy-induced,
definition of, 243 therapy in, 498-497 182
with ethanol injection anesthesia in, 498, 481 bioheat equation of, 27, 30,
therapy, 304-305,473 clinical applications of, 206,208,286-287
as hepatocellular carcinoma 498-504 evaluation of extent of, 112
treatment, 6, 218, 243, 322, historical perspective on, 498 Coagulation testing, prior to
323,326-327,473 of liver cancer, 499, 501-502 ablation procedures, 440
with laser-induced of lung cancer, 502, 503 Coagulopathy,
thermotherapy, 245 of osteoid osteoma, 498-481 cryotherapy-related, 446,
as liver cancer treatment, pain associated with, 357, 447
243 498-499 Collagen denaturation, in
with radiofrequency ablation of renal cancer, 499, 502, 504 laser-induced interstitial
therapy, 304-305 spinal anesthesia thermotherapy, 235
Chemotherapy. See also specific contraindication in, 395 Collateral injury, to organs
chemotherapeutic agents Cholangiocarcinoma, imaging of, adjacent to tumors, 108
antiangiogenic effects of, 48 127 prevention of, 108, 109
deposition in hypervascular Chondroblastoma, differentiated Colon
tissue, 297 from osteoid osteoma, ablation-related damage to,
electroporation-assisted, 295 389,393 108
for liver metastases, 243-244, Chondroma, differentiated from liver tumors adjacent to, 487
482-483 osteoid osteoma, 392-393 radiofrequency
metastatic colorectal Chondrosarcoma, 492-515 ablation-related
cancer-related, 303, 467, Chordoma, 287 perforation of, 485
482-483 Chronic obstructive pulmonary Colorectal cancer, metastatic
with radiofrequency disease (COPD), 502 chemotherapy for, 467,
ablation, 315 Cilengitide, 47 482-483
Index 521

contrast-enhanced ultrasound pulmonary metastasectomy comparison with ultrasound,


imaging of, 143 of, 354 97-98
hepatic metastatic, 242-243 radiofrequency ablation of, of hepatocellular carcinoma
ablative therapy for, 466-468 113,114,214 ablation therapy, 327, 473,
chemotherapy for, 303, 467, radiofrequency ablation of 475
482-483 with CT guidance, 139 of liver metastases ablation
combination therapies for, in the liver, 287, 315-318, therapy, 314-315
269-270 466-467 of lung cancer ablation
contraindications to in the lung, 113, 114, 214 therapy, 357-358, 359, 360,
percutaneous therapy for, survival rates in, 486 361
59 with ultrasound guidance, multislice, 104
cryotherapy for, 11, 101, 154, 139 technique of, 105-106
156,467,486 recurrent, 483 patient's comments about, 512
curative ablation of, surgical treatment for, postablation hypoattenuating
466-467,468 242-243,311,466,482,483 areas in, 112
ethanol injection therapy cancer recurrence following, preoperative, prior to
for, 243 483 radiofrequency ablation,
extrahepatic disease survival rates in, 482-483, 486 210
associated with, 487 tumor ablation of, 483 of radiofrequency ablation,
intraoperative treatment for, unresectable, 487 115-117
59 Combretastatin,47 contrast-enhanced patterns
laser-induced interstitial Complications, of ablation in, 115
thermotherapy for, 237, procedures, 440-439. See for hepatic lesions, 115-117
243,244-245 also specific complications for hepatocellular
microwave coagulation of cryotherapy, 261-262 carcinoma, 473, 475
therapy for, 230-231, 232 CT imaging of, 116 for lung cancer, 357-358,
palliative ablation therapy of uterine artery embolization, 359,360,361
for, 467-468 417-418 for osteoid osteoma, 390,
patient/family narratives Computed tomography (CT), 391,392
about, 506-513 104-120 residual active tumor foci
postablation follow-up comparison with nuclear on, 115-116
imaging of, 114 medicine imaging, 121 real-time, 104
postablation scar volume in, gas bubble distribution in, 112 technique of, 105
114 helical, 311 of renal cell carcinoma, 343,
prevalence of, 234, 482 contrast-enhanced, of 344,347,348,350-351
radiofrequency ablation of, hepatic lesions, 145 Computed tomography guidance,
287,315-318,466-467 multidetector, of liver 104
recurrent, 483 metastases, 313 axial images in, 109-110
surgical treatment for, multiphasic contrast agent use in, 109-110
242-243,311,466,482, contrast-enhanced, of of cryotherapy, 25, 154,
483 liver metastases, 136 257-259,263,447
survival rates in, 482-483 Computed tomography electromagnetic targeting
metastatic to rib, vertebral fluoroscopy, 184-185, 209, systems in, 106-107
body, and pleural surface, 258-259 of ethanol injection therapy,
383,384 Computed tomography 157,473
positron emission tomography follow-up, 104, 114-118 of extremities tumor ablation
of, 125 of breast cancer/tumor therapy, 106
prevalence of, 482 ablation therapy, 115 indications for, 104-105
pulmonary metastatic of ethanol injection therapy, laser goniometers in, 106, 107
CT-monitored 199,200,473,475 of laser-induced interstitial
radiofrequency ablation helical contrast-enhanced, 145, thermotherapy, 159,238,
of, 113, 114 314-315 240
patient's personal narrative of hepatic ablation therapy, of liver cancer/tumor ablation,
about, 514-515 115-117,145 104-105,107-108,110,159
522 Index

Computed tomography guidance Cryodestruction, 154 CT imaging of, 259


(cant.) Cryoshock,261-262,446-447 "hot spots" within, 188
comparison with ultrasound, Cryosurgery, 154 monitoring of, 251-252
104-105 Cryotherapy, 182,250-272,369, MR imaging of, 260
of microwave coagulation 403-404 in prostate cryotherapy, 254
therapy, 475 action mechanisms of, 25-26, real-time imaging of, 187
in overlapping ablations, 110 154,252-253,422 ultrasound imaging of,
of radiofrequency ablation argon gas-based systems, 25, 100-101,254,255,256,257
in children, 481 83-84,252,253-254,484 immunologic response to,
of hepatic abscesses, 116 Joule-Thompson effect in, 422-423
of hepatocellular carcinoma, 83 intracellular ice formation in,
475,476 argon/helium gas-based 252,422
image fusion in, 292 systems, 84-86 Joule-Thompson effect in, 83,
patient's comments about, Endocare system, 84-86 423
491 Galil system, 84, 85 laparoscopic, 256
in pediatric lung cancer for breast cancer/tumors, 154, liquid nitrogen-based, 25,
patients, 502 422-427,428,436-438 250-251,253,254,484
of retroperitoneal biopsies, 107 as cryoprobe-assisted for liver tumors, 154
robotic, 290 lumpectomy, 425 complications of, 446-448
technique in, 105-112 of fibroadenomas, 422-425 extrahepatic disease as
Computed tomography as primary therapy, 425-426 contraindication to, 486
monitoring, 104, 112-114 as cell death/damage cause, history of, 251
Congestive heart failure, renal 182,422,436 laparoscopic, 482
cryotherapy-related, 449 for colorectal cancer-related rninilaparotomies in, 256
Conscious sedation, 68-69 hepatic metastases, 467 ultrasound guidance of,
Constipation, uterine comparison with other ablative 254-257
leiomyoma-related, 413 techniques, 228, 261-263 for lung cancer, 257
Contrast agent allergy, as complications of, 11, 60, for metastatic colorectal
contraindication to 261-262 cancer, 11, 101, 154, 156,
uterine artery cryogens in, 484 467,486
embolization, 414 history of, 250 for metastatic neuroendocrine
Contrast agents cryoprobes in, 25 tumors, 334-335, 468-469
gadolinium, 117,237,238, MRI-compatible, 259 for metastatic rectal cancer,
239-240,278-279,280,313 placement of, 99-100 373
hepatobiliary-specific,311 size of, 99 minimum lethal temperature
liver-specific, 313 ~-guided,25, 154,257-259, in, 26
reticuloendothelial-specific, 259,263,447 MRI-guided, 25, 154, 156,
311,313 definition of, 154 259-261,263
temperature-sensitive, 151-152 double-freezing technique in, for bone tumors, 156
tumor-seeking, 184 243 comparison with
use in computed tomography, drug depot effect of, 294, 297 laser-induced interstitial
109-110 equipment for, 253-254 thermotherapy, 243
Control, of image-guided tumor extracellular ice formation patients' comments about,
ablation therapy, 149, during, 252 515-491,491-492
188-189 gas bubbles associated with, for prostate cancer, 408,
Control groups, 18 116-117 409
Cooled-wet technique, 30, 31 heating blanket use during, 447 rationale for, 156
Cordotomy, 167 for hepatocellular carcinoma, for rectal cancer/tumors,
Coronary artery disease 326,486 158-159
screening, prior to high-pressure gas systems in, for renal cancer/tumors, 156,
interventional MRI 423 186
procedures, 153 history of, 11,250-252,484 for soft tissue tumors, 156
Cough, radiofrequency iceballs in, 251-252 MRI monitoring of, 187
ablation-related, 462-463 acoustic shadowing of, 100 open surgical, 446
in children, 502 in argon gas systems, 254 pain associated with, 68
Index 523

palliative, for metastatic soft D E


tissue tumors, 371, 372, Dana-Farber Cancer Institute, Electrical impedance
373 508,511,512,514,491, tomography (EIT), 296
of presacral rectal tumor, 372 494-496 Electromagnetic targeting
for prostate cancer DCI01,49 systems, 106-107
complications of, 251, Denonvilliers' fascia, saline Electroporation, 295-296
448-449 injections into, 407 Embolism, pulmonary
conglomerate iceballs in, Des-gamma--carboxy-prothromb cryotherapy-related, 447,448
254 in, as hepatocellular uterine artery
with CT guidance, 408 carcinoma marker, 199, embolization-related, 418
history of, 251 327 Embolization, transarterial
Medicare reimbursement Diabetes mellitus, 443 (TAE), 468-450, See also
for, 63 Diaphragm Balloon arterial occlusion;
with MRI guidance, 408, hepatocellular carcinoma Chemoembolization
409 adjacent to, 476 of the hepatic artery
as nerve-sparing therapy, hepatocellular carcinomas for hepatocellular
405-406 underneath, 226 carcinoma, 322, 326-327
patient selection for, radiofrequency for neuroendocrine
403-406 ablation-related injury to, metastatic tumors, 334
with saline injection into 487 prior to radiofrequency
Denonvilliers' fascia, 407 radiofrequency ablation, 209
as salvage therapy, 406-407, ablation-related paralysis of the uterine artery, 276, 277,
448 of, 485 412-421
technical advances in, Dielectrics, of tissue, 287 as adenomyosis treatment,
407-409 Disseminated intravascular 413
with ultrasound guidance, coagulation, complications of, 417-418
402-411 cryotherapy-related, as fibroid disease treatment,
for renal cancer, 156, 175-176, 446 412-421
186 Dosimetry, 185, 189 future directions in, 418
complications of, 176, Doxil,35-36 historical background of,
449-450 Doxorubicin, 6, 35-36, 304, 312, 412
skin protection during, 373 468,484,495 indications for, 412-413
thermal variables in, 436 Doxorubicin-eluting polymer outcomes of, 417-418
tumor seeding associated with, implants, 295 patient selection for,
491-492 Drilling system, for bone tumor 413-414
ultrasound-guided, 154, 251, access, 110, 111 technique in, 414-417
254-257,263 Droperidol, contraindication in Emphysema, 355-356
artifacts in, 254-257 Parkinson's disease, 356 Endocare cryoablation
for breast lesions, 423, 424, Drug delivery system,84-86
425,436 targeted, 167 Endometrial carcinoma,
for liver metastases, 154, ultrasound-mediated, 296 metastatic, 381, 382
156 via liposomes/nanoparticles, Endostatin, 45, 47
for needle and cryoprobe 35-36,297-298 Endothelial cells, in tumor
placement, 99-100 via radiofrequency ablation, vascularization, 43-44
CT. See Computed tomography 294 Enzinger, P. c., 509
C-225,509 Drug depot effect, 294-295, 297 Epidemiology, clinical, 17-22
Current Procedural Terminology Drug depots, polymerized, 287, noncomparative studies in,
(CPT) codes, 63, 74 288 18-22
Cystic artery, in uterine artery Dyspareunia, Epidermal growth factor
embolization, 414 leiomyoma-related, 413 receptor inhibitors, 483
Cysts Dysphonia, ethanol injection Equipment, for tumor ablation,
hepatic, 98 therapy-related, 442 288. See also Instruments
renal, 9 Dyspnea, renal Erythema, necrolytic migratory,
Cytokines, cryotherapy-related cryotherapy-related, 336
release of, 446-447 449 Esophageal cancer, 244
524 Index

Ethanol injection therapy, for liver metastases, 24 Fibroadenoma


195-204 comparison with cryotherapy for, 423-425, 436,
action mechanism of, 24, 25 laser-induced interstitial 437-438
complications of, 44Q-442 thermotherapy, 243 focused high-intensity
CT-guided, 157 in liver transplantation ultrasound therapy for,
diameter of coagulation candidates, 202 160-161
necrosis in, 24 for osteoid osteoma, 394 spontaneous regression of,
efficacy of, 24 pain associated with, 68 422
for hepatocellular carcinoma, as palliative therapy Fibroids, uterine. See
9,156-157,322-324,325, in head and neck cancer Leiomyoma, uterine
328 patients, 370 Fibrosis, idiopathic pulmonary,
action mechanism of, 195 in liver tumor patients, 195 356
adverse effects and for painful bone metastases, FISP (fast imaging with
complications of, 198 378 steady-state precession
with chemoembolization, prior to radiofrequency techniques), 172-173
266-267,473 ablation, 374 Fistulas
comparison with with radiofrequency ablation, arterioportal, 115
laser-induced interstitial 302 rectourethral, prostate
thermotherapy, 243 in children, 502 cryotherapy-related, 448,
contraindications to, for retroperitoneal tumors, 371 449
198-199 as tumor necrosis cause, 68 rectovesical, prostate
with contrast-enhanced CT ultrasound-guided, 157 cryotherapy-related,
imaging, 112 needle placement in, 99, 100 449
conventional technique in, volume of injected ethanol in, Flumazenil, 68
195,196-197 286 Fluorine-18-fluoro-2-deoxy-D-
CT-guided, 473 Etiomidate, 69 glucose (FDG). See
equipment for, 195-196 Extremities, tumors of, 373 Positron emission
indications for, 202 CT-guided ablation of, 106 tomography (PET), with
inflammatory tissue rim in, recurrent, 371 fluorine-18-fluoro-2-deox
115 y-D-glucose (FDG)
internists' performance of, F 5-Fluorodeoxyuridine, 483
472-474 Families, of tumor ablation Fluoroscopy
limitations to, 218, 243 patients, 506-516 computed tomographic (CT),
mortality rate in, 198, 201 Fast imaging with steady-state 184-185,258-259
multiple-needle insertion precession techniques x-ray, 184-185
technique in, 473 (FISP), 172-173 5-Fluorouracil, 269, 303, 467, 468,
principles of, 195 Fat, perirenal, 117, 118 482,509,514
procedure for, 195 Femoral nerve, radiofrequency Focused high-intensity
rationale for use of, ablation-related injury to, ultrasound therapy, 182,
201-202 371,372 273-284,369
single-session technique in, Femur ablation volume in, 286
197-198,199 eosinophilic granuloma of, action mechanism of, 34, 160
survival rates in, 473-474 393 advantages and disadvantages
therapeutic efficacy osteoid osteoma of, 390, 391 of, 34, 275-276
evaluation of, 199-201 Fentanyl, 68, 69, 356-357 areas of coagulation necrosis
tumor seeding in, 198 a-Fetoprotein, 199, 327, 486 in, 34
ultrasound monitoring of, Fever, postprocedural, 441 for breast cancer/tumors,
196 radiofrequency 160-161,428,440-436
history of, 9 ablation-related, 314, 443, comparison with laser-induced
limitations to, 24-25 444,485 interstitial thermotherapy,
for liver cancer/tumors, 9 in children, 502 243
complications of, 108, uterine artery definition of, 160
44Q-442 embolization-related, drug depot effect of, 297
as palliative therapy, 195 417 history of, 12, 273
Index 525

image guidance applications of G H


comparison with computed Gadolinium, as MRI contrast Halo pattern, of recurrent
tomography, 104-105 agent, 117,237,238, hepatocellular carcinoma,
in hepatic tumors, 104-105 239-240,278-279,280, 115-116
InSightec-TxSonics system, 313 Head and neck cancer
89 Galil cryotherapy system, 84, ablation treatment for, 370
intraoperative, 184-185 85 bleomycin
in cryotherapy, 100-101 Gallbladder electrochemotherapy for,
of focal liver masses, 313 ablation-related collateral 295
for liver cancer assessment, damage to, 108 cryotherapy for, 154
487 liver lesions/tumors adjacent laser-induced interstitial
for liver metastases to, 210,487,501-502 thermotherapy for, 10, 245
detection, 60, 136 Gas bubbles, 116-117 in recurrent cancer, 240-241,
laparoscopic, 226 Gastric cancer 244
for liver tumors, 12 ablative therapy for, 466 pulmonary metastasectomy
as liver tumor treatment, metastatic, 371 treatment for, 354
12 ablative therapy for, 469 Heat efficacy, 206
MRI-guided, 190,275-276 laser-induced interstitial Heat sink effect, 6, 114, 116, 286,
advantages of, 190 thermotherapy for, 244 301,302,502
as breast tumor treatment, MR imaging of, 128 Heat transfer, in thermal
160-161 positron emission ablation therapy, 182,
as leiomyoma treatment, tomography of, 128 286-287
160,161,276-282 radiofrequency ablation of, Heimlich valves, 357
MRI-monitored, 243 318 Helium, as cryoprobe, 423
noninvasiveness of, 190 Gastric metastases, Hematoma, subcapsular, 245,475
operation of, 89 leiomyosarcoma-related, Hemobilia,474
physical principles of 175 Hemolysis, 198, 485
beam focusing, 274 Gastrinoma, 333 Hemoperitoneum, 198, 442, 475
propagation through tissue, Gastrointestinal cancer. See also Hemoptysis, 48
273-274 Colorectal cancer; Gastric Hemorrhage
tissue effects, 274-275 cancer abnormal vaginal,
for prostate cancer, 12 ablative therapy for, 466 leiomyoma-related,
for renal cancer, 175-176 radiofrequency ablation of, 412-313
sonoporation effect of, 296 486 cryotherapy-related, 262, 447,
targeting applications of, 149 Gastrointestinal stromal tumors 449
thermal insensitivity of, 185 (GIST), 128-129, 150, ethanol injection
use in gene therapy, 293 208 therapy-related, 198,441,
for uterine leiomyomas Gelfoam,33 474
of atypical hypercellular Gemcitabine,469-470 intraabdominal
leiomyomas, 280 General anesthesia, 67, 69, 357 laser-induced interstitial
MRI-guided,276-282 Gene therapy, 167,292-293, thermotherapy-related,
patient preparation for, 295-296,484 245
281 Genetronics, Inc., 295 microwave coagulation
treatment planning in, Glioma, cerebral, 184 therapy-related, 224
278-280 Glucagonoma, 333, 336 microwave coagulation
treatment procedure in, Goniometers, laser, 106, 107 therapy-related, 224
282 Granulation tissue, postablation, peritoneal, 441, 474
Folinic acid (leucovorin), 303, 115 radiofrequency
482,483 Granuloma, eosinophilic, 393 ablation-related, 262, 485
Food and Drug Administration Groin, sarcoma of, 372 hepatic, 485
(FDA), 485 "Growing pains," 373-390 intraperitoneal, 476
Fractures, stress, 393 Gynecologic cancer/tumors. See pulmonary, 117, 443, 444,
Free-hand technique, 99, 100 also Leiomyoma, uterine 462,463
Fuchs, Charles, 514 hepatic metastatic, 318 renal, 176
526 Index

Hemorrhage (cant.) with microwave coagulation procedure for, 195


subscapsular, microwave therapy, 268-269 rationale for use of, 201-202
coagulation preoperative, 267, 268 single-session technique in,
therapy-related, 224 chemoprevention of, 305 197-198
umbilical cord chemotherapy for, 484 survival rates in, 473--474
compression-related, in children, 501 therapeutic efficacy
272 cirrhosis associated with, 472, evaluation of,199-201,
Hemothorax,224,441,442,475 483--484 202
Hepatectomy, 318, 334, 468 ethanol injection therapy tumor seeding in, 198
Hepatic artery for, 24, 200 experimental treatment for,
chemoembolization of radiofrequency ablation of, 484
as hepatocellular carcinoma 209,211 fibrous capsule of, 218
treatment, 6, 468, 484 ultrasound imaging of, hepatectomy treatment for,
in liver transplantation 141-142 468
recipients, 468 ultrasound screening for, 201 hepatitis B-associated, 209,
as chemotherapeutic agent combination therapies for, 211,484
infusion site, 467--468, 483, 266-272 hepatitis C-associated, 484
484 neoadjuvant therapy, hot saline injection therapy
embolization of 266-268 for, 10
as neuroendocrine contrast-enhanced CT imaging immunoprevention of, 305
metastatic tumor of,138 incidence of, 483
treatment, 334, 468--469 cryotherapy for, 326, 486 internists' treatment of,
prior to radiofrequency diagnosis of, 322 472--481
ablation, 209 embolization of, 322, 326-327 intraoperative ultrasound
occlusion of, 209, 269, 301 ethanol injection therapy for, detection of, 98
balloon occlusion, 6, 32-33, 9,156-157,195,322-324, intratumoral injection therapy
209,301 325,328 for, 30
with Pringle maneuver, 6, action mechanism of, 195 laser-induced interstitial
101-102,209,212,213 adverse effects and thermotherapy for, 326
Hepatic vein complications of, 198 with chemoembolization,
balloon occlusion of, 312 with chemoembolization, 269
in ethanol injection therapy, 266-267,473 liver transplantation treatment
197 comparison with for, 23,266,267,322,328,
radiofrequency laser-induced interstitial 468,472
ablation-related injury to, thermotherapy, 243 microwave coagulation
476 complications of, 108 therapy for, 11,230-231,
Hepatitis, 135 contraindications to, 198-199 322,325-326
Hepatitis B, 209, 211, 484 conventional technique in, adverse effects and
Hepatitis C, 484 195,196-197 complications of,
Hepatoblastoma, 501, 502 cost of, 202 224-225
Hepatocellular carcinoma, 23, CT-guided,112,473 with chemoembolization,
322-331 equipment for, 195-196 268-269
acetic acid injection therapy indications for, 202 comparison with
for, 324 inflammatory tissue rim in, radiofrequency ablation,
chemoembolization of, 6, 218, 115 225,475,480
304-305,322,323, internists' performance of, complications of, 475
326-327,328,468,472, 472--474 CT assessment of, 475
473,484 limitations to, 218, 243 with embolization, 223-224
in children, 501 local tumor recurrence after, internists' performance of,
with ethanol injection 201 474--475,480
therapy, 266-267, 473 mortality rates in, 198, 201 laparoscopic, 225-226
with laser-induced multiple-needle insertion limitations to, 480
interstitial thermotherapy, technique in, 473 local cancer recurrence
269 principles of, 195 after, 223
Index 527

microwave delivery system surgical resection of, 218, 322, thermotherapy;


in, 474 328,484 Radiofrequency
percutaneous, 218-226 chemoembolization prior to, ablation
results of, 222-223 267,268 limitations to monitoring of,
survival rates in, 222-223 eligibility for, 210 101
ultrasound-guided, 475 recurrence following, 472 lIypotension,
MR imaging of, 129 survival rate in, 210 cryotherapy-related,
multistep modality tumor markers for, 327 446
image-guided local ultrasound-guided ablation of, IIypothermia,
treatment for, 327-328 218 cryotherapy-related,447
positron emission tomography ultrasound imaging of, lIypoxemia, radiofrequency
of, 129 137-138,140-142 ablation-related, 443
prevalence of, 472, 483 underneath the diaphragm, 226 lIysterectomy, 276, 414, 418
radiofrequency ablation of, lIepatoma, 135,269
135,200,323,324-325, lIerceptin, 46--47 I
328,475,482 lIigh-temperature ablation. See Ibuprofen, 390
artificial pleural effusion lIyperthermic ablation Ifosfamide, 495
technique in, 476 lIilum, hepatic, lesions adjacent Ileus, paralytic, 449, 450
comparison with ethanol to, 210 Image-guidance, of tumor
injection therapy, 480 lIippocrates, 154 ablation therapy, 23--40.
comparison with microwave lIormonal therapy, for liver See also Computed
coagulation therapy, 225, metastases, 315 tomography (CT);
475,480 lIounsfield units (lIU), 110,259 Magnetic resonance
CT assessment of, 475, 476 5-lIydroxyindoleacetic acid imaging (MRI); Positron
CT-guided, 476 as carcinoid syndrome marker, emission tomography
first use of, 159 333 (PET); Ultrasound
internists' performance of, as carcinoid tumor marker, 338 limitations to, 190
475--479 lIypertension principles of, 23-24, 183-189
laparoscopic, 302 pheochromocytoma-related, control, 149, 188-189
local tumor recurrence in, 337 interactive
476 radiofrequency localization/targeting,
MRI-guided, 173 ablation-related, 443, 445 184-185
"oven effect" in, 311-312 IIyperthermia monitoring, 149, 185,
postablation scar volume in, cytotoxic levels of, 484--485 187-188
114 effect on drug uptake, 296 planning, 148
size of coagulation necrosis effect on vascular targeting, 148-149, 184-185,
in, 210, 211 permeability, 296-297 186
survival rates in, 468, 486 laser-induced interstitial tumor margin localization,
therapeutic efficacy of, 475 therapy-induced, 10, 157, 183-184
tissue dielectrics in, 287 159 real-time, 184-185
tumor seeding associated low-temperature, 293 theory of, 23-36
with, 443 malignant, 66 Imaging and Therapeutic
ultrasound-guided, microwave therapy-induced, Technology, 507
137-145 10 Immunotherapy, 293
with vascular occlusion radiofrequency Impedance, 76
techniques, 6, 301 ablation-related, 50 Impotence, prostate
recurrent effect of chemotherapy on, cryotherapy-related, 448
CT detection of, 116 35-36,304 Infarction
halo pattern of, 115-116 synergistic interaction with hepatic, 224
screening for, in Japan, 480 radiation, 293 renal, 117-118
staging of, 322 lIyperthermic ablation, 26. See segmental, 485
subscapular, 210, 211 also Focused Infection
tumor seeding associated high-intensity ultrasound; ablation procedures-related,
with, 110 Laser-induced interstitial 440
528 Index

Infection (cant.) L MRI monitoring of, 234


pelvic, as contraindication to Laparoscopy MR thermometry monitoring
uterine artery in cryotherapy, 446, 482 of, 236-238
embolization, 413-414 in intraoperative operation of, 86-88
uterine artery ultrasound-guided optical fiber use in, 182, 190
embolization-related,418 procedures, 95, 97-98 for osteoid osteoma, 110,394
Inferior vena cava in microwave coagulation for painful bone metastases,
hepatocellular carcinoma therapy, 225-226 378
adjacent to, 479 in radiofrequency ablation, PhotoMedex system in, 86-89
metastases to, 213 212-213,482 pull-back technique in, 236
Infertility, uterine Laparotomy scattering dome applicator in,
leiomyoma-related, 413 in cryotherapy, 243 235-236
Inflammatory tissue rim, as hepatic tumor treatment, for soft tissue tumors, 242
postablation, 115, 116 446 temperature-dependent effects
InSightec-TxSonics focused as leiomyoma treatment, 414 of, 235
high-intensity ultrasound open, 302 thermal mapping in, 188
ablation system, 89 Laplace's equation, 296 ultrasound-guided, 159
Institutional review board (IRB), Laser goniometers, 106, 107 needle placement in, 99
59 Laser-induced interstitial ultrasound monitoring of, 234
Instruments thermotherapy (UTI), Lavamisol,514
ferromagnetic, 172 157,159,234-249,369 Leiomyoma, uterine
MRI--<:ompatible, 153-154, 209 ablation volume in, 190,286 cryotherapy for, 276
Insulinoma, 333 action mechanism of, 34, 234, focused high-intensity
Intercostal nerve blocks, 371 235 ultrasound treatment for,
Interferon-a/~, antiangiogenic application systems and 160,161,276
activity of, 47 techniques in, 235-236 of atypical hypercellular
Internet, 61-62 for breast tumors, 159,440 leiomyomas, 280
Internists, hepatocellular comparison with MRI-guided, 276-282
carcinoma treatment by, radiofrequency ablation, patient preparation for, 281
472-481 190 treatment planning in,
Iressa (ZD1839), 46-47, 514 CT-guided, 159 278-280
Irinotecan, 467, 483, 509, 514 device modifications in, 34 treatment procedure in, 282
Islet cell tumors, 332, 333, 469 energy dose delivery in, 189 hysterectomy treatment for,
for hepatocellular carcinoma, 276
J 326 laser-induced interstitial
Jaklitch, Michael, 514, 515 with chemoembolization, ablation of, 159,276
Japan, hepatocellular carcinoma 269 myomectomy treatment for,
treatment in, 472-481 history of, 10 276,413
Jaundice, radiofrequency for leiomyoma, 159,276 partial ablation of, 183
ablation-related, 476 for liver cancer/tumors, 238 uterine artery embolization
Joint effusion, osteoid complications of, 450-451 treatment for, 276, 277,
osteoma-related, 392 CT-guided, 159 412-421
Joule-Thompson effect, 83, 423 MRI-guided, 159 complications of,418
Jugular vein, thrombosis of, 441 ultrasound-guided, 159 future directions in, 418
for liver metastases historical background of, 412
K clinical studies of, 244-245 indications for, 412-413
Katzen, Barry, 60 comparison with alternative outcomes of, 417-418
Kidney, radiofrequency methods, 242-244 patient selection for,
ablation-related tumor complications of, 245 413-414
seeding in, 445 indications for, 242 technique in, 414-417
Kidney cancer. See Renal in vitro studies of, 244 Leiomyosarcoma
cancer qualitative and quantitative in children, 501
Kidney tumors. See Renal cell evaluation of, 240, 242 metastatic, 175,373
carcinoma MRI-guided, 159, 234-249 recurrent, 371
Index 529

Leucovorin, 467 minilaparotomies in, 256 small animal model of,


Leukotomy, 167 ultrasound-guided, 254-257 173-174
Levovist, 136 CT-guided ablation of, transthoracic approach in,
Limb paralysis, radiofrequency 107-108,110,257,259 487
ablation-related, 371, 372 comparison with ultrasound, ultrasound-guided, 485
Lipiodol, 33, 327, 476, 484 104-105 recurrent, following surgical
Liposomes, use in drug delivery, ethanol injection therapy for, 9 resection, 210
35-36,297-298 complications of, 108, surgical resection of, 103,
Liquid nitrogen, use as cryogen, 440-442 210
25,250-251,253,254,484 as palliative therapy, 195 tumor margin localization in,
LITT. See Laser-induced focal, in children, 502 183
interstitial thermotherapy focused high-intensity ultrasound imaging of
Liver ultrasound treatment for, intraoperative, 97-98
abscess of 12 limitations of, 135
cryotherapy-related, intraoperative ultrasound Liver decompensation, ethanol
447-448 imaging of, 487 injection-related, 198
ethanol injection laser-induced interstitial Liver disease, bilobar, 334
therapy-related, 441 thermotherapy for, 238 Liver failure
microwave coagulation complications of, 450-451 cryotherapy-related, 261, 446,
therapy-related, 451, 475 CT-guided, 159 447
radiofrequency MRI-guided, 159 ethanol injection
ablation-related, 442, 476, ultrasound-guided, 159 therapy-related, 441
485,499 microwave ablation of, laser-induced interstitial
infarction of 228-233 thermotherapy-related,
ethanol injection in animal models, 229, 230 450
therapy-related, 474 of large-volume tumors, microwave coagulation
microwave coagulation 228-233 therapy-related, 451
therapy-related, 224 MRI-guided, 162 radiofrequency
radiofrequency new applicators in, 229-233 ablation-related, 442, 476,
ablation-related, 476 radiofrequency ablation of, 4 485
radiofrequency antibiotic prophylaxis in, 499 Liver lesions. See also Liver
ablation-related tumor basic technique in, 485 cancer/tumors; Liver
seeding in, 443 blood flow reduction during, metastases
sonographic hyperechoic areas 10 combination radiofrequency
in, 112 in children, 499, 501-502 ablation/chemotherapy
Liver Cancer Study Group of complications of, 442-443 treatment of, 35-36
Japan, 323 contraindications to, 485 ultrasound imaging of, 135-147
Liver cancer/tumors factors affecting the efficacy for assessment of treatment
ablation of, anesthesia during, of, 174 results, 137-145
68 FDA approval for, 485 with contrast-enhanced
adjacent to the gallbladder, generator systems in, 485 ultrasound, 135-147
501-502 impedance-based, 485 Liver metastases, 311-321
biopsies of laparoscopic approach in, adjacent to the gastric wall,
free-hand technique in, 99 212-213,482,485,486-487 109
ultrasound-guided, 98-99 MRI-guided,173-175 breast cancer-related, 115,212,
comparison with renal tumors, open surgical approach in, 303
341 212-213,486-487 intraoperative ultrasound
cryotherapy for, 154 outcomes of, 486 detection of, 98
complications of, 446-448 output-based, 485 radiofrequency ablation of,
CT-guided, 257, 259 patient's personal narrative 318
extrahepatic disease as about, 513-514 chemoembolization of
contraindication to, 486 percutaneous approach in, comparison with
history of, 251 211-212,486-487 laser-induced interstitial
laparoscopic, 482 with Pringle maneuver, 485 thermotherapy, 243
530 Index

Liver metastases (cant.) comparison with colorectal cancer-related,


following surgical resection, laser-induced interstitial 315-318
269-270 thermotherapy, 243 comparison with
with radiofrequency ethanol injection therapy for, laser-induced interstitial
ablation, 303, 304 24 thermotherapy, 243
chemotherapy for, 243-244, comparison with comparison with surgical
303,482--483 laser-induced interstitial metastasectomy,311
comparison with thermotherapy, 243 contraindications to, 312-313
laser-induced interstitial focused, high-intensity in the hepatic dome, 169
thermotherapy, 243-244 ultrasound therapy for, with hormonal therapy, 315
colorectal cancer-related 243 indications for, 135, 312
ablative therapy for, gastrointestinal stromal in metastatic
466--468,483 cancer-related, 150 neuroendocrine tumors,
chemotherapy for, 303, 467, hepatectomy treatment of, 318-319
482--483 318 MRI-guided, 173-175
combination therapies for, imaging of, 313 pain management in, 314
269-270 intraoperative ultrasound patients' comments about,
contraindications to detection of, 136 491--491
percutaneous treatment laser-induced interstitial patient selection for,
of,60 thermotherapy for, 312-313
cryotherapy for, 11, 101, 234-238 patients' narratives about,
154,156,467,486 clinical studies of, 244-245 491--491
curative ablation of, comparison with alternative percutaneous approach in,
466--467,468 methods, 242-244 211-212,486--487
ethanol injection therapy complications of, 245 procedure in, 313-315
for, 243 indications for, 242 safety margin in, 312
extrahepatic disease in vitro studies of, 244 tissue dielectrics in, 287
associated with, 487 qualitative and quantitative surgical resection of
intraoperative treatment for, evaluation of, 240, 242 comparison with
60 localization of, 184 laser-induced interstitial
laser-induced interstitial metachronous, 312, 317 thermotherapy, 242-243
thermotherapy for, 237, MRI-guided biopsy of, 150 eligibility for, 210
243,244-245 neuroendocrine tumor-related survival rate in, 210
microwave coagulation cryotherapy for, 469 ultrasound detection of,
ablation of, 230-231, 232 hepatic artery embolization sensitivity of, 136
palliative ablation therapy of,469 Liver transplantation, as
for, 467--468 radiofrequency ablation of, hepatocellular carcinoma
patient/family narratives 318,469 treatment, 23, 266, 267,
about, 506-513 surgical treatment for, 322,328,468,472
postablation follow-up 333-334 Liver transplantation patients
imaging of, 114 parenchymal involvement in, ethanol injection therapy in,
postablation scar volume in, 210-211 202
114 pathology of, 311-312 hepatic arterial
prevalence of, 234, 482 patients' and patients' families' chemoembolization in,
radiofrequency ablative narratives about, 506--491 468
therapy for, 287, 315-318, radiofrequency ablation of, Lobectomy, as lung cancer
466--467 159-160,209,270 treatment, 358, 360
recurrent, 483 advantages of,311 Long bones, MRI-guided
surgical resection of, 466, area of coagulation necrosis radiofrequency ablation
482,483 in, 312 in, 177
survival rate in, 482--483 breast cancer-related, 318 Low back pain, uterine
contrast-enhanced MRI of, with chemotherapy, 315 fibroids-related,413
184 coagulation necrosis margins Lower extremities, osteoid
cryotherapy for, 11, 155,467 in, 210-211 osteoma of, 392
Index 531

Lumpectomy, cryoprobe- patient preparation for, electrodes in, 171


assisted, 425 356 of the female pelvis, 276-277
Lung cancer potential applications of, interventionai, 209
combination therapy for, 353, 357-362 artifacts in, 153
459-460,461 for primary lung cancer, aspiration applications of,
conventional treatment for, 357-361,461-463 170
459-460 sedation during, 356-357 biopsy applications of,
cryotherapy for, CT-guided, in single-lung patients, 355 170
257 technique in, 356-357 C-arm system for, 172
CT-guided ablation of, 111, recurrent, 358,360 closed systems, 170
257 small cell, 353 disadvantages of, 170
metastatic surgical resection of, 459, 461 history of, 170
ablation of, 370--371 cancer recurrence rate image acquisition times in,
to bone and soft tissue, 371, following, 358, 360 170
377 symptoms of, 354 open systems, 170
in children, 502, 503 Lung metastases. See also Lung scanners in, 170--171, 172
isolated, 361-362 cancer, metastatic suite design for, 170--172
locally recurrent, 463 breast cancer-related, 312 temperature-sensitive
palliative treatment for, 355 colorectal cancer-related techniques in, 151, 152,
radiation therapy for, 354 CT-monitored 173,185,275-276
radiofrequency ablation of, radiofrequency ablation temporal resolution in,
209,362,363,364,365, of, 113, 114 170
463-464 patient's narrative about, intraoperative, 184-185
surgical treatment for, 354, 514-515 irreversible tissue necrosis
361-362 pulmonary metastasectomy delineation with, 188
mortality rate in, 353, 354 of,354 in laser-induced interstitial
non-small-cell' 353 radiofrequency ablation of, thermotherapy (UTI),
radiofrequency ablation of, 113,114,214 234,236-248
209,214 ethanol injection of liver lesions, comparison
pain associated with, 354-355 therapy-related, 441 with ultrasound, 97-98
palliative ablative therapy for, radiofrequency ablation of, of osteoid osteoma, 392
353,354-355,358,362, 209,362,363 patient's comments about,
370-371,502,503 renal cell carcinoma-related, 511-512
positron emission tomography 362 preoperative, prior to
(PET) of, 125-127 Lymph nodes, cervical, metastatic radiofrequency ablation,
prognosis for, 353 disease of, 370 210
radiation therapy for, 353, 354 Lymphoma, 287 pulse sequence development
radiofrequency ablation of, in, 178
168,209,214,353-368 M rapid gradient echo pulse
action mechanism of, Magnetic resonance imaging sequences in, 172
353-354 (MRI) of renal cell carcinoma, 343,
animal model of,460-461 advantages of, 167, 170 344
in children, 502, 503 in combination with positron role of, in thermal ablation
complications of, 214, emission tomography, 125, therapy, 188
443-444 126,127,128 safety issues of, 172-173
contraindications to, 356, 357 comparison with nuclear scanner-related acoustic noise
early clinical experiences medicine imaging, 121 generated during, 172
with,357-362 contrast-enhanced temperature mapping with. See
for metachronous lung with gadolinium, 237, 238, Magnetic resonance
cancer, 461-462 239-240,280,313 thermometry
as outpatient procedure, liver- and temperature-sensitive, 151,
357 reticuloendothelial-specifi 152,173, 185
as palliative therapy, 358, c,313 in focused ultrasound
362,502,503 of liver metastases, 184 surgery, 275-276
532 Index

Magnetic resonance imaging open systems, 152 short-tau inversion recovery


follow-up scanners in, 152 (STIR) sequences in,
contrast-enhanced, of liver suites for, 152-153 174-175,177
metastases ablation, targeting applications of, 151 system grounding in, 173
314-315 temperature-sensitive temperature-sensitivity of,
of hepatocellular carcinoma contrast agents in, 173
ablation therapy, 327 151-152 turbo spin echo imaging in,
of liver lesions ablation temperature-sensitive 173
therapy, 115, 116 mapping methods in, turbo spin echo sequences
Magnetic resonance imaging 151-152 in, 174-175
guidance, 148-166 of laser-induced interstitial in vertebrae, 177
advantages of, 149-152 thermotherapy (LITI), targeting applications of, 149
of biopsies, 149-151 159,234-249 technical advances in, 148
comparison with in breast cancer treatment, temperature-sensitive
computed tomography, 104, 440,436 mapping in. See Magnetic
149 for liver cancer/tumors, 159 resonance thermometry
ultrasound,149 of liver metastases biopsies, triangulation method in, 149
of cryotherapy, 25, 154, 156, 150 Magnetic resonance imaging
259-261,263 of microwave interventional suites,
for bone tumors, 156 thermocoagulation, 162 hazards in, 172
comparison with planning component of, 149 Magnetic resonance
laser-induced interstitial of prostate brachytherapy, 162, thermometry, 112, 171,
thermotherapy, 243 163 289
patients' comments about, of prostate gland biopsies, 149, advantages of, 170
515-491,491-492 151 diffusion coefficient-based
for prostate cancer, 408, 409 of radiofrequency ablation, 160 methods, 151, 152
rationale for, 156 for abdominal masses, 160 fast low-angle shot (FLASH),
as rectal cancer/tumor advances in, 170-172 238,240,241,244
treatment, 158-159 in animal models, 173-174, in laser-induced interstitial
as renal cancedtumor 176 thermotherapy, 236-238
treatment, 156, 186 artifacts in, 173 proton resonance frequency
as soft tissue tumor for cancer, 167-181 (PRF) method, 151, 152
treatment, 156 comparison with surgical Tl-based methods, 151, 152
of ethanol injection therapy, approach, 173 Mammography, 151
157 contrast enhanced sequences Massachusetts General Hospital,
of focused high-intensity in, 174-175 514
ultrasound therapy, cost-effectiveness of, 173 Matrix metalloproteinase
160-161,190,275-276 fast imaging with inhibitors, as cancer
advantages of, 190 steady-state precession treatment, 45
as breast tumor treatment, techniques (FISP), Matrix metalloproteinases, in
160-161 172-173 tumor angiogenesis, 42-43
as leiomyoma treatment, for liver cancer/tumors, McMullen, William, 511
160,161,276-282 173-175 Media exposure, 62-63
interventional, 148-154 in long bones, 177 Mediastinum, CT-guided biopsy
closed systems, 152 over- or underablation in, of, 107
history and development of, 173 Medicare, 63, 74
152-154 for pancreatic cancer, 177 MEDLINE,18
interventional device pulse sequences used for, Memorial Sloan-Kettering
tracking systems of, 151 172-173 Cancer Center, 491, 495
monitoring applications of, renal, 175-177 Mesothelioma, metastatic, 371,
151-152 for renal cancer, 175-177 462
MRI-eompatible research and future Metastasectomy
instrumentation for, developments in, 177-178 hepatic, 319
153-154 safety issues in, 172-173 pulmonary, 354, 360
Index 533

Metastases. See also specific survival rates in, 222-223 Myoma, uterine. See Leiomyoma,
types of metastases, e.g., ultrasound-guided, 475 uterine
Bladder metastases history of, 11 Myomectomy, 276, 414
angiogenesis in, 44-45 laparoscopic, for effect on fertility, 413
Methotrexate, 495 hepatocellular carcinoma,
Meyerhardt, Jeffrey, 514 225-226 N
Miami Vascular Institute, 60 of liver tumors, 228-233 Naloxone, 68
Microbubbles, 101 in animal models, 229, 230 Nanoparticles, 298
Microspheres, as uterine artery of large-volume tumors, Naproxen, 390
embolization agents, 416 228-233 Narcotics
Microstreaming, 296 MRI-guided,162 use as conscious sedation, 68
Microtaze AZM-520 MW new applicators in, use as monitored anesthesia
ablation system, 88 229-233 care (MAC), 69
Microthrombosis, ethanol mechanisms of, 219-220 National Blue Cross/Blue Shield
injection therapy-related, methods in, 220-222 Technology Assessment
198 new applicators in, 229, 230 Committee, 62
Microwave coagulation ablation power output assessment in, Necrosis, ablation
therapy,11,228-233,369 219-220 therapy-related, 261, 262
ablation volume in, 286 probes in, 33-34 cryotherapy-related, 261, 262
action mechanism of, 162, 228 for renal cancer, 175-176 comparison with
advantages of, 228 results of, 222-223 radiofrequency ablation,
adverse effects and survival rates in, 222-223 262-263
complications of, systems operation in,88 radiofrequency
224-225 tumor-specific immune ablation-related, 261,
comparison with response to, 293 262-263
cryotherapy, 228 ultrasound-guided needle size of, 114
radiofrequency ablation, placement in, 99 Nephrectomy, as renal cell
225,228 Microwave generators, 219 carcinoma treatment,
direct field heating in, 228 Microwave ovens, 228 342-343
electrodes in, 33, 219, 220 Midazolam, 68, 69, 356-357 Nerve injuries
with embolization, 223-224 Mitomycin C, 269, 484 patient positioning-related,
endometrial, 229 Modeling, in tumor ablation 73-74
for hepatocellular carcinoma, therapy, 286-287 radiofrequency
218-226,230-231,322, Monitored anesthesia care ablation-related, 371, 372,
325-326,475 (MAC),69 373
adverse effects and Monitoring, in image-guided Neuroendocrine tumors
complications of, 224-225 tumor ablation therapy, combination therapy for, 303
with chemoembolization, 149,185,187-188 hormonal hypersecretion
268-269 MRI. See Magnetic resonance associated with, 469
comparison with imaging metastatic, 332-340
radiofrequency ablation, Mueller, Peter, 510 ablative therapy for, 468-469
225,475,480 Mueller, Susan, 510 chemotherapy for, 334-335,
complications of,475 Multiorgan failure 468-469
CT assessment of, 475 cryotherapy-related, 446, 447 cryotherapy for, 258,
with embolization, 223-224 laser-induced interstitial 334-335
internists' performance of, thermotherapy-related, embolization of, 334
474-475,480 450 radiofrequency ablation of,
laparoscopic, 225-226 Myocardial infarction, 315-318,335-339
limitations to, 480 cryotherapy-related, 447 surgical treatment for,
local cancer recurrence Myocardial ischemia, in patients 333-334
after, 223 undergoing interventional microwave ablation of, 230
microwave delivery system MRI,153 New technologies, in tumor
in,474 Myoglobinuria, cryotherapy- ablation, 285-300
results of, 222-223 related,447 Nidus, 390, 392
534 Index

NM-3,47 pain associated with, postprocedural


Nocturia, uterine 498-499 in liver metastases patients,
fibroids-related,413 patient preparation for, 314
Noncomparative studies, of 395-396 in uterine artery
ablation therapy, 18-22 postprocedure patient care embolization patients, 417
Nonrandomized trials in, 398 with radiofrequency ablation,
control groups in, 18 preoperative imaging, 167,213,378-386,494-515
evaluation of, 18-22 390-392 PAKY-RCM robotic arm, 290
excluded patients in, 19 success rates in, 399 Palliative ablative therapy
inception cohorts in, 18-19 spontaneous resolution of, 394 for colorectal cancer-related
outcomes assessment in, 20-22 Osteosarcoma, intracortical hepatic metastases,
endpoints, 20 variant of, 393 467-468
quality of life outcome, Ovarian arteries, in uterine effect on survival, 21
21-22 artery embolization, 417 ethanol injections as
response to therapy Ovarian cancer, metastatic, 127 for head and neck cancer
outcome, 20 Ovary, in uterine artery patients, 370
survival outcome, 21 embolization, 416-417 for liver tumor patients,
time to progression "Oven effect," 295-312 195
outcome, 20-21 Oxaliplatin, 303, 467, 484, 514 for lung cancer, 353, 354-355,
patient entry criteria in, 19 358,362,370-371,502,503
patient selection in, 18-19 p for metastastic disease of bone
selection bias in, 18 Pacemakers, hepatic and soft tissue, 371, 372,
statistical considerations in, radiofrequency 373,377-388
22 ablation-related for neuroendocrine hepatic
study protocols in, 20 dysfunction of, 485 metastases, 334, 335
Nonsteroidal anti-inflammatory Pain radiofrequency ablation as,
drugs, 390, 394 ablation therapy-related, 167,213
Nuclear medicine imaging, 121 mechanisms of, 67-68 in bone metastases patients,
alcohol reflux-related, 196 378-386
o ethanol injection in lung cancer patients, 362
Occupational Safety and Health therapy-related, 196, 198 in metastatic colorectal
Administration (OSHA), lung cancer-related, 354-355 cancer patients, 467-468
72 osteoid osteoma-related, 213 in pediatric liver tumor
Oil, ethiodized. See Lipiodol radiofrequency patients, 502
Oncocytoma, management of, ablation-related in pediatric lung tumor
345-346 in children, 498-499 patients, 502, 503
Osteitis pubis, prostate in lung cancer patients, 357 personal narrative about,
cryotherapy-related, 449 Pain management. See also 494-515
Osteoblastoma, differential Anesthesia; Palliative in soft tissue tumor patients,
diagnosis of, 389 ablative therapy 369,370
Osteoid osteoma of bone metastases/bone Pallidotomy, 167
CT-guided ablation of, 110 tumor-related pain Pancoast tumors, as pain cause,
definition of, 389, 499 computed tomography in, 354-355
differentiated diagnosis of, 389, 110-111 Pancreatic cancer
392-393 with ethanol injections, 378 clinical course of, 468
laser-induced interstitial with laser-induced metastatic
thermotherapy for, 110 interstitial thermotherapy, chemotherapy for, 469-470
pain management in, 213 378 laser-induced
radiofrequency ablation of, with radiofrequency thermotherapy for, 244
209,213,369,389-401 ablation, 378-386 MRI-guided radiofrequency
in children, 498-481 in lung cancer patients, ablation of, 177
contraindications to, 394-395 370-371 Pancreaticoduodenectomy
equipment for, 396-398 in osteoid osteoma patients, (Whipple resection),
follow-up of, 398-399 390 469
Index 535

Pancreatitis, ablation therapy- microwave coagulation radiofrequency ablation-


related, 371, 447, 449 therapy-related, 224, 451, related, 442, 476, 485
Paralysis, radiofrequency 475 Positron emission tomography
ablation-related, 371, 372, as radiofrequency ablation (PET), 121-136
373 contraindication, 357 in breast cancer ablation
Parkinson's disease, as radiofrequency therapy follow-up, 438
droperidol ablation-related, 117, 214, with
contraindication, 356 443,444,462-463 fluorine-18-fluoro-2-deox
Patients, comments about tumor Pleural surface, metastatic y-D-glucose (FDG),
ablation therapy, 506-515, lesions of, 383, 384 121-134,315
506-516 Pleurisy in cholangiocarcinoma, 127
PElT. See Ethanol injection as radiofrequency ablation clinical considerations in,
therapy, percutaneous contraindication, 357 123-124
Pelvic mass, as contraindication radiofrequency in colorectal cancer, 125, 126
to uterine artery ablation-related, 462-463 with computed tomography,
embolization, 413-414 Pleuritic pain, radiofrequency 124,127,129
Pelvic tumors, cryotherapy for, ablation-related, 502 FDG biodistribution in,
251 Pneumonectomy, 462 122-123
Pelvis metastatic lung cancer in gastric cancer, 128
CT-guided biopsy of, 107 following, 365 in gastrointestinal stromal
female, MRI imaging of, Pneumothorax tumors (GIST), 128-129
276-277 ablation therapy-related, 73 in hepatocellular carcinoma,
Percutaneous tumor ablation ethanol injection 129
therapy. See also specific therapy-related, 108 in lung cancer, 125-127, 128
therapies induced, in liver tumor with magnetic resonance
comparison with ablation, 108 imaging, 125, 126, 127, 128
intraoperative tumor in lung cancer patients, 357 methodology of, 123
ablation, 102 microwave coagulation in ovarian cancer, 127
Peritoneal metastases, gastric therapy-related, 451 patient preparation for, 122
cancer-related, 469 postablation, chest tube pharmacokinetics of, 122
Peritonitis placement in, 111 principles of, 121-122
laser-induced interstitial as radiofrequency ablation in renal cell carcinoma, 129
thermotherapy-related, contraindication, 357 in hepatocellular carcinoma
450 radiofrequency ablation of, 108 ablation therapy
radiofrequency radiofrequency follow-up, 327
ablation-related, 442, 476 ablation-related, 113, 117, in lung cancer, 357-358
PET. See Positron emission 214,443,444,462,485 scanners in, 292
tomography Polyprenoic acid, 305 Postablation syndrome, 349,
Pharyngeal cancer, 244 Polyvinyl alcohol particles, as 443
Phase transitions, uterine artery in children, 499
thermally-induced, 188 embolization agents, Postembolization syndrome, 417
Pheochromocytoma, metastatic, 414-416 Practice building, in
337 Portal vein image-guided tumor
Photocoagulation, interstitial occlusion of, 32-33, 301 ablation therapy, 59-63
laser. See Laser-induced with balloon occlusion, 442 Pregnancy
interstitial thermotherapy with Pringle maneuver, 6, after uterine artery
PhotoMedex laser ablation 32-33,101-102,209,212, embolization, 413
system, 86-88 213 as contraindication to uterine
Planning, in image-guided tumor thrombosis of artery embolization,
ablation therapy, 148 cryotherapy-related, 447 413-414
Pleural effusion ethanol injection intrauterine radiofrequency
laser-induced interstitial therapy-related, 441 ablation during, 445
thermotherapy-related, microwave coagulation Pringle maneuver, 6, 32-33,
245,450 therapy-related, 451 101-102,209,212,213
536 Index

Probes rectal cancer invasion of, 373 optimal temperature for, 8


end-fire endorectal prostate, thermal ablation-related action mechanisms of, 3, 26-27,
99 injuries to, 289 159,205,428-429
energy dose delivery through, transuretheal resection of, as adjuvant therapies with,
189-190 urethral sloughing 207-208
for intraoperative ultrasound, treatment, 449 of adrenal lesions,
95-96 Prostate-specific antigen, 149, complications of, 445-446
laparoscopic, 96-97, 99 151,404 anesthesia during, 68
microwave, 33-34 Proteinases, in tumor antiangiogenic effects of, 50
technological development of, angiogenesis, 42-43 as biliary tract thermal injury
288 Protein denaturation, thermal cause, 210
temperature-sensitive, 185 ablation-related, 182, 235, bioheat equation of, 206, 207
thermal diffusion from, 182 261 bipolar arrays in, 28
ultrasound, optimal trajectory Proteins, tumor-derived, role in blood flow-induced tissue
for, 185, 186 angiogenesis, 41-42 cooling during, 207,
Procrit, 491, 492 Prothrombin time, as ethanol 208-209
Propofol, 69 injection therapy blood flow modulation during,
Prostaglandins, production in contraindication, 198-199 6,30,32-33,209,301
bone tumors, 389 Pubic symphysis, metastatic with electrode overlapping,
Prostate cancer lesions of, 382 312
brachytherapy for Pulmonary disease patients, with mechanical occlusion,
MRI-guided, 162, 163 anesthesia in, 66 32-33,312
ultrasound-guided,161-162 Pyothorax, microwave with pharmacologic
cryotherapy for, 154 coagulation modification, 33, 312
complications of, 251, therapy-related, 475 with Pringle maneuver, 212,
448-449 213
conglomerate iceballs in, 254 Q of bony metastases, 209
CT-guided, 408 Quality of life, effect of tumor complications of, 445
history of, 251 ablation on, 21-22 of brain metastases, 209
Medicare reimbursement of breast cancer/tumors, 168,
for, 63 R 428-440
MRI-guided, 408, 409 Radiation, synergistic effects with chemotherapy, 303
as nerve-sparing therapy, with hyperthermia, 293 complications of, 446
405-406 Radiation therapy. See also equipment for, 430
patient selection for, Brachytherapy sonographic guidance of,
403-406 in combination with 429-438,439
with saline injection into radiofrequency ablation, burn variability of, 287, 288
Denonvilliers' fascia, 407 213 with chemotherapy, 8
as salvage therapy, 406-407, external-beam in children, 498-497
448 for bone pain, 377 anesthesia in, 498
technical advances in, for lung cancer, 353, 354 clinical applications of,
407-409 for recurrent lung cancer, 498-504
ultrasound-guided, 402-411 360 historical perspective on, 498
focused high-intensity palliative, for lung cancer, 355 of liver cancer, 499, 501-502
ultrasound treatment for, planning stage in, 185 of lung cancer, 502, 503
12 radiofrequency ablation as of osteoid osteoma, 498-481
metastatic to bone, 377 alternative to, 213 pain associated with,
thermal ablation of, tumor Radiofrequency ablation, 498-499
margins in, 183 205-217 ofrenalcancer,499,502,504
Prostatectomy, comparison with ablation volume (tissue coagulation necrosis in, 26-27
prostate cryotherapy, necrosis) in, 3-4, 5, 6-7, blood flow reduction-related
402-403,404,406 8-9,286 increase in, 32-33
Prostate gland effect of percutaneous perfusion-mediated
biopsy of, 149, 151 needles on,S, 6 decrease in, 32
Index 537

saline infusion-related internally-cooled,28-29, with hypotensive anesthesia,


increase in, 31-32 207,502 301-302
temperature requirements nonperfused,168-169 image fusion in, 292
for, 304 perfused, 30 inadequate tissue coagulation
of colorectal cancer water-cooled,169-170 in, 207-208
metastases, 46~67 extrahepatic, 213-214, 341 blood flow-related, 207,
CT-guided, 139 foil grounding pad use in, 499 208-209,210
survival rate in, 467, 468 general principles of, 167-173 intrauterine, during pregnancy,
with ultrasound guidance, goal of, 205 445
139 as groin tumor treatment, laparoscopic techniques in, 302
in combination with 372 ultrasound-guided, 212
balloon occlusion, 287-288 grounding pads in, 3, 4 limitations of, 243
brachytherapy, 361 heat efficacy of, 206 of liver cancer/tumors, 168,
chemoembolization, 209, heat sink effect in, 6, 211, 301, 482--490,487
287-288 302 antibiotic prophylaxis for,
chemotherapy, 287, 303-304 in pediatric lung cancer 499
ethanol injection therapy, in patients, 502 basic technique in, 485
children, 502 of hepatocellular carcinoma, blood flow reduction during,
intratumoral injections, 302 135,200,209,323, 8
radiation therapy, 213 324--325,328,475,482 in children, 501-502
surgical resection, 302 artificial pleural effusion complications of, 485
comparison with technique in, 476 contraindications to, 485
cryotherapy, 261, 262-263 comparison with ethanol FDA approval for, 485
laser-induced interstitial injection therapy, 480 generator systems in, 485
thermotherapy, 190,243 comparison with microwave impedance-based, 485
microwave ablation, 228 coagulation therapy, 225, laparoscopic approach in,
complications of, 60, 262, 475,480 212-213,482,485,486--487
442--444 complications of,476 MRI-guided, 173-175
in children, 499 CT assessment of, 475, 476 open surgical approach in,
in lung cancer patients, CT-guided,476 212-213,48~87
462--463 first use of, 159 outcomes of, 486
nerve injuries, 371, 372, 373 internists' performance of, output-based,485
paralysis, 371, 372, 373 475--479 patient's personal narrative
in pediatric lung cancer laparoscopic, 302 about, 513-514
patients, 502 local tumor recurrence in, percutaneous approach in,
contraindications to, in lung 476 211-212,48~87
cancer patients, 357 MRI-guided, 173 with Pringle maneuver, 485
with contrast-enhanced "oven effect" in, 311-312 small animal model of,
ultrasound, 137 postablation scar volume in, 173-174
cooled-wet technique in, 30, 31 114 transthoracic approach in,
CT-guided, 209 . size of coagulation necrosis 487
in children, 481 in, 210, 211 ultrasound-guided, 485
as debulking therapy, 294, 298 survival rates in, 468 of liver lesions
definition of, 159 therapeutic efficacy of, 475 complications of, 442--443
drug delivery systems and, tissue dielectrics in, 287 efficacy evaluation of,
294 tumor seeding associated 115-117
liposome-based, 32-33, with,443 surgeon's perspective on,
297-298 ultrasound-guided,137-145 482--490
drug depot effect of, 294--295, with vascular occlusion of liver metastases, 159-160,
297 techniques, 6, 301 209,270
efficacy of, 168 history of, 3-9, 484--485 advantages of, 311
electrodes in, 77 as hyperthermia cause, 50 area of coagulation necrosis
clustered internally-cooled, hypervascular tissue in, 294, in, 312
29 297 breast cancer-related, 318
538 Index

Radiofrequency ablation (cont.) MRI-guided, 160, 168-178, for painful bone metastases
with chemotherapy, 315 190,209 treatment, 378-386
coagulation necrosis margins of abdominal masses, 160 as palliative therapy, 213
in, 210-211, 212 electrode tip temperature in, in Beckwith-Wiedemann
colorectal cancer-related, 168-169,170 syndrome, 502
315-318 evaluation phase in, 170 in pediatric liver tumor
comparison with surgical follow-up visualization in, patients, 502
metastasectomy,311 170 in pediatric lung cancer
contraindications to, 312-313 guidance phase in, 168,169 patients, 502, 503
in the hepatic dome, 169 of liver cancer, 174 personal narrative about,
with hormonal therapy, 315 in long bones, 177 494-515
indications for, 135, 312-313 of pancreatic cancer, 177 in renal cell carcinoma
MRI-guided, 173-175 planning phase in, 168 treatment, 214
neuroendocrine renal, 175-177 of pancreatic tumors, 168
tumors-related, 318-319 of renal cancer, 175-177 patient eligibility criteria for,
pain management in, 314 research and future 210
patient's comments about, developments in, 177-178 perfusion effect in, 287
491-491 treatment phase in, 168 physiologic alterations of
patient selection for, in vertebrae, 177 tissue in, 30-33
312-313 MRI-monitored, fast thermal with altered tissue thermal
percutaneous approach in, technique in, 187 and electrical conductivity,
211-212,486-487 multiple-array needles in, 31-32
procedure in, 313-315 109 with continuous saline
safety margins in, 312 multiple probe arrays in, 27-28 infusions, 30-32
tissue dielectrics in, 287 needles used in, 3-4 with improved tissue heat
of lung cancer/tumors, 168, design of, 3-4, 6, 7 conduction, 31
209,214,353-368 needle track ablation in, 207 preoperative imaging studies
action mechanism of, of neuroendocrine hepatic in, 210
353-354 metastases, 332, 335-339 probes in, 160
animal model of, 460-461 of non-small-celliung cancer, of prostate tumors, 168
in children, 502, 503 209 pulmonary
complications of, 214, operation of, 76-83 complications of, 443-444
443-444 Berchtold system, 77-'f1,2 CT monitoring of, 113-114
contraindications to, 356, 357 Boston Scientific system, lung tissue density in,
early clinical experiences 79-80 113-114
with, 357-362 Celon system,82-83 postablation imaging of, 113,
of metachronous lung grounding pads in, 75 117
cancer, 461-462 in monopolar-type systems, pulsed, 29-30
as outpatient procedure, 76-77 as radiation therapy
357 Radionics system, 75-79 alternative, 213
as palliative therapy, 358, RITA Medical Systems, Inc. of renal cancer/tumors, 168
362,502,503 system, 80-82 in children, 502, 504
patient preparation for, 356 of osteoid osteoma, 209, 213 of renal cell carcinoma, 209,
potential applications of, in children, 498-481 343-351
357-362 contraindications to, 394-395 needle biopsy prior to,
of primary lung cancer, equipment for, 396-398 345-346
357-361,461-463 follow-up of, 398-399 patient selection for,
sedation during, 356-357 patient preparation for, 343-344
in single-lung patients, 355 395-396 planning of, 344-345
technique in, 356-357 postprocedure patient care of renal lesions, complications
magnetic needle tracking in, 398 of, 444-445
systems in, 290-292 success rates in, 399 repeated applications of, 168
as metastatic neuroendocrine pain management applications retractable needle electrode
tumor treatment, 468-469 of, 167,213 systems of, 206-207
Index 539

of retroperitoneal Radionics radiofrequency complications of, 349


metastases/tumors, 209, ablation needles, 7 MRI-guided,176-177
371 Radionics radiofrequency needle biopsy prior to,
robotics-assisted, 290-290 ablation system, 77-79 345-346
. safety of, 168 Randomized controlled trials, 18 as palliative therapy, 214
saline infusion-enhanced, 6, Rectal cancer patient selection for,
208,213,302 metastatic, to the prostate 343-344
of soft tissue tumors, 369 gland,373 saline-enhanced, 213
of splenic tumors, 168 MRI-guided cryotherapy for, staging of, 341
sublethal, effect on gene 158-159 surgical resection of, 341,
transfection, 293 recurrent, 379 342-343
synergism with radiotherapy, presacral,371,372,381-382 von Rippel-Lindau
293 Referred pain, osteoid disease-associated, 502,
as thermal damage cause, osteoma-related, 392 504
288-289 Regional anesthesia, 69 Renal collecting system
thermal lesion shape and size Reimbursement, for anesthesia ablation-related thermal injury
in, 167-168 services, 74-75 to,445
thermal mapping in, 188 Renal cancer in cryotherapy, 449-450
thermistor monitoring of, 168 cryotherapy for, 156, 175-176, Renal failure. See Liver failure
thermometry-guided, 186 Renal lesions, radiofrequency
288-289 complications of, 176, ablation of, 444-445
of thyroid cancer, 370 449-450 Renal tumors
tissue dielectrics in, 287 focused high-intensity ablation of
tissue necrosis in ultrasound ablation of, anesthesia during, 68
effect of arterial occlusion 175-176 tissue dielectrics in, 287
on, 301 microwave ablation of, cryotherapy for
effect of blood flow 175-176 CT-guided, 257
reduction on, 301 radiofrequency ablation of MRI-guided, 156, 157, 186
tissue temperature during, 205, animal model of, 176 postablation CT-based
484-485 in children, 499, 502, 504 evaluation of, 117-118
tissue temperature monitoring clinical trial of, 176-177 Response Evaluation Criteria in
devices in, 206-207 MRI-guided,175-177 Solid Tumors (RECIST),
transpleurodiaphragmatic, of Renal cell carcinoma, 341-352, 114
pneumothoraces, 108 371 Retroperitoneal tumors, 209, 371
tumor environment in, 287-288 cryotherapy for, MRI-guided, Retroperitoneum, CT-guided
tumor-free margins in, 157,260 biopsy of, 107
205-206 grading of, 341-342 Rib, metastatic lesions of, 383,
tumor modeling in, 287 imaging of 384
tumor-specific immune during ablation, 347, 348 RITA Medical System
response to, 293 postablation, 349-351 radiofrequency ablation
ultrasound-guided, 5, 209 preablation, 344-345 needles, 6
of breast cancer, 429-438, metastatic RITA Medical Systems, Inc.
439 to the brain, 363 radiofrequency ablation
echogenic response in, 5 to the kidneys, 363 system, 80-82
of hepatocellular carcinoma, to the lungs, 362, 363 Robotics, 289-290
159 pulmonary metastasectomy Rofecoxib, 47
of liver metastases, 313-314 in, 354 Ropivacaine,314
for needle placement, 99 positron emission tomography Rosen, Gerald, 495
Radiologists, 489 of,129
tumor ablation practices of, prognosis of, 341-342 S
59--63 radiofrequency ablation of, Sacrum
getting started, 59-61 209,213-214,343-351 fractures of, as pain cause, 381
growing the practice, 61-63 ablation volume in, 288 metastatic lesions of, 383-384,
payment issues in, 63 clinical experiences in, 351 385
540 Index

Sacrum (cont.) in the extremities, 371, 373 Survival, effect of tumor ablation
radiofrequency ablation of, in the head and neck, 370 on, 21-22
380-383 indications for, 370 Swanson, Richard, 514
rectal carcinoma-related, as palliative therapy, 370
381-383 in the pelvis, 371, 372 T
recurrent rectal carcinoma special situations in, TACE. See Chemoembolization,
anterior to, 381, 382 373-374 transarterial
St. Vincent's Cancer Center, 495 in the thorax, 370-371 TAE. See Embolization,
Saline infusion definition of, 369 transarterial
into Denonviliers' fascia, 407 laser-induced interstitial Targeting, in image-guided
percutaneous hot, 10 thermotherapy for, 242 tumor ablation therapy,
as protective adjunctive MRI-guided cryotherapy for, 148-149,184-185, 186
technique, 373 156 Teratoma, sacrococcygeal, 445
radiofrequency radiofrequency ablation of, Testicular cancer, hepatic
ablation-enhancing, 6, 484 metastases from, 244, 318
208,213,302 SOMATEX laser application kit, Thalamotomy, 167
Sarcoma 238 Thalidomide, 47
of the extremities, 371, 373 Somatic pain, 67 Thermal ablation therapies, 167,
osteogenic, 354 Somatostatin analogues, 303, 182. See also Focused
soft tissue, 354 468-469,469 high-intensity ultrasound
Science News, 509 Sonoporation, 296 therapy; Laser-induced
Scoliosis, spinal osteoid SonoVue, 136, 143,144 interstitial thermotherapy;
osteoma-related, 394 Spinal anesthesia, 69 Microwave coagulation
Sedation, in radiofrequency Spinal cord metastases, 68, 213 ablation therapy;
ablation patients Spinal cord tumor ablation, Radiofrequency ablation
in lung cancer patients, anesthesia during, 68 ablation volume in, 182-183
356-357 Splanchnic nerves, 67 action mechanism of, 261
as nausea and gastric Squamous cell carcinoma with adjuvant chemotherapy,
aspiration cause, 356 laser-induced thermotherapy 5-36
for renal cell carcinoma for, 245 bioheat equation of, 27, 30,
ablation, 346 recurrent parastomal, 241 206,208,286-287
Segmentectomy, as lung cancer Stenting, ureteral, preprocedural, blood flow in, effect on
treatment, 360 373 ablation volume, 183
Selection bias, 18 Stents energy deposition control in,
Sepsis, pulmonary, 117 biliary, 447 182-183
Shankar, Sridhar, 510-511, 513, retrograde ureteral, 344 heat delivery in, 27
514,494-495,496 Stereotactic guidance, of heat transfer mechanisms in,
Shock, septic, 450 laser-induced interstitial 182-183
Shock waves, 296 thermotherapy, 440 invasiveness of, 182
Shunt, arterioportal, 451 Stereotactic neurosurgery, 167 limitations to, 190
Sildenafil, 448 Stomach planning stage in, 185
Skeletal metastases. See Bone ablation-related collateral real-time MRI monitoring of,
metastases damage to, 108, 109 190
Skin, ablation-related injury to. liver tumors adjacent to, 487 temperature requirements in,
See also Burns Stress fractures, 393 185
prevention of, 373 Subscapular hemorrhage, thermal gradients in, 189-190
Small bowel tumors, 469 microwave coagulation tissue margins in, 182
Soft tissue and bone lesions, therapy-related, 224 Thermistors, 168
369 Subscapular metastases, direct (dependent), 289
Soft tissue tumors postablation CT imaging indirect (independent), 289
ablation of, 369-376 of, 115 Thermometry, 288-276. See also
in the abdomen, 371 Surgeons, perspective on hepatic Magnetic resonance
adjunctive procedures in, radiofrequency ablation, thermometry
373-374 482-490 Thiopental, 69
Index 541

Thoracic neoplasms. See Lung Tumor ablation therapy, contrast-enhanced


cancer image-guided advantages of, 135
Thoracoabdominal tumors, ablation volume in, 112 continuous-mode, hepatic
cryotherapy for, 251 building a practice in, 59-63 applications of, 136-137
Thoracotomy, 362 clinical epidemiology of, 17-22 hepatic, with radiofrequency
Thrombocytopenia, 261, 446, 485 noncomparative studies, ablation, 137
Thrombosis 18-22 for liver metastases
of the jugular vein, 442 differentiated from biopsies, detection, 136, 313
of the portal vein, 451 108-109 microbubble contrast agents
cryotherapy-related,447 efficacy evaluation of, 114 in, 136, 140
ethanol injection in the kidneys, 117-118 principle of, 136
therapy-related, 441 in the liver, 115-117 conventional unenhanced, 135,
radiofrequency in the lungs, 117 136
ablation-related, 442 general principles of, 285-288 definition of, 273
vascular, 261, 262 history of, 3-16, 484-485 .Doppler
vascular endothelial growth incomplete, follow-up imaging contrast-enhanced harmonic
factor pathway of,114 power, 116
inhibitors-related, 48 limitations of, 285 with ethanol injection
Thumb-printing method, in new technologies in, 285-300 therapy, 196, 197
thermal ablation pain associated with, 67-68 for hepatocellular carcinoma
monitoring, 185, 188 patients' and their families' ablation follow-up,
Thyroid cancer, 370, 469 comments about, 506-516 137-138,140-142
Thyroid nodules, ethanol thermal lesion size and shape for hepatocellular carcinoma
injection treatment for, in, 285, 286 therapy evaluation, 327
442 toxicity of, 22 of liver tumors, limitations to,
Tissue ablation, definition of, Tumor growth, 135
76 angiogenesis-dependent. Ultrasound follow-up
TNP-478,50 See Angiogenesis, in contrast-enhanced
Transitional cell cancer, 371 tumor growth of ethanol injection therapy,
Trazodone, 509 Tumor margin localization, 199,200
Triangulation method, 149 183-184 of liver metastases ablation,
Tumor ablation systems Tumor necrosis factor, 21 315
operation, 76-90 Tumor seeding of hepatocellular carcinoma
applicators in, 76 cryotherapy-related, 491-492 ablation, 137-138, 140-142
of cryotherapy systems, 83-86 ethanol injection Ultrasound guidance
feedback in, 76 therapy-related, 198,441, of acetic acid injections, 10
of focused ultrasound ablation 474 contrast-enhanced, of hepatic
systems, 89-91 liver tumor ablation-related, ablation therapy, 137-145
of laser-induced interstitial 110 of cryotherapy, 154,251,
ablation systems, 86-88 microwave coagulation 254-257,263
main control panel in, 76 therapy-related, 224, 451 artifacts in, 254-257, 257
of microwave coagulation multiple ablation needle for breast lesions, 423, 424,
ablation systems, 88 punctures-related, 109 425,436
parameters and settings in, radiofrequency for liver metastases, 154,
76 ablation-related, 442, 443, 156
of radiofrequency ablation 445,476 for needle and cryoprobe
systems Tumstatin,47 placement,99-100
Berchtold system, 77-82 Tyrosine kinase inhibitors, 483 of ethanol injection therapy, 9,
Boston Scientific system, 99,100,157,475
79-80 U of extremity tumor ablation,
Celon system, 82-82 Ultrasound,135-147 106
Radionics system, 77-79 advantages of, 137 intraoperative (IOUS)
RITA Medical Systems, Inc. B-mode, for liver metastases equipment for, 95-97
system, 80-82 detection, 313 laparoscopic, 95, 97-98
542 Index

laparoscopic probes in, of percutaneous laser therapy, monoclonal antibodies against,


96-97 234 49
of liver tumors, 97-98 Ultrasound screening, for Vascular endothelial growth
probe covers for, 95-97 hepatocellular carcinoma, factor (VEGF) expression
probes for, 95 322 vector, 46
procedures in,95-103 Upper respiratory infections, as Vascular endothelial growth
scanning technique in, contraindication to factor (VEGF) pathway
97-98 anesthesia, 66 antagonists, 46, 48-49
ultrasound guidance Ureters, cryotherapy-related Vascular endothelial growth
techniques in, 98-102 injury to, 253 factor (VEGF) receptor
of intratumoral chemotherapy, Urethra, cryotherapy-related inhibitors, 484
9-10 injury to, 253 Vasoactive intestinal
of laser-induced interstitial Urethral sloughing, prostate peptide-secreting tumors
thermotherapy (LITI), cryotherapy-related, (VIPomas),333
99, 159 449 Vasoconstriction, thermal
of liver biopsies, 98-99 Urinary frequency, uterine ablation-related, 188
of microwave coagulation fibroids-related,413 Vasodilatation, thermal
therapy, 475 Urinary incontinence, prostate ablation-related, 188
of prostate brachytherapy, cryotherapy-related, 448 Ventricular fibrillation, 153
161-162 Urologists, involvement in renal Vertebrae, MRI-guided
of prostate cryotherapy, cell carcinoma radiofrequency ablation
402-411 radiofrequency ablation, in, 177
in nerve-sparing therapy, 344 Vertebral bodies, metastatic
405-406 Uterine artery, anatomy of, lesions of, 383, 384
patient selection for, 414 VIPomas (vasoactive intestinal
403-406 Uterine artery embolization, as peptide-secreting tumors),
with saline injection into uterine leiomyoma 333
Denonvilliers' fascia, 407 treatment, 276, 277, Visceral pain, 67
as salvage therapy, 406-407 412-421 Vitaxin, 46, 47
technical advances in, complications of,417-418 von Hippel-Lindau disease, 175,
407-409 future directions in, 418 502,504
of radiofrequency ablation historical background of, 412
in hepatocellular carcinoma indications for, 412-413 W
treatment, 159 outcomes of, 417-418 Water, interaction with
with laparoscopic approach, patient selection for, 413-414 microwave irradiation,
212 technique in, 414-417 219,228
of liver metastases, 313-314 Uterine myoma. See Leiomyoma, Whipple resection
in pediatric lung cancer uterine (pancreaticoduodenectom
patients, 502 y),469
in renal cell carcinoma V World Health Organization,
treatment, 213 vanSonnenberg, Eric, 507, 508, 114
transrectal, 149, 151 509,510,512-514,496
Ultrasound-mediated particle Vaporization, of tissue, X
delivery systems, 296 radiofrequency X-rays, of osteoid osteoma, 391
Ultrasound monitoring ablation-related, 205, 206 X-ray therapy, palliative, for
comparison with CT Varices, bleeding, 441 bone metastastic cancer,
monitoring, 112 Vascular endothelial growth 355
of laser-induced interstitial factor (VEGF)
thermotherapy (LITI), in angiogenesis, 41-42, 43, 45, Z
234 50 Zoloft,509

You might also like