You are on page 1of 349

Handbook of

Radioembolization
IMAGING IN MEDICAL DIAGNOSIS AND THERAPY
Series Editors: Andrew Karellas and Bruce R. Thomadsen

Published titles

Quality and Safety in Radiotherapy Physics of Thermal Therapy:


Todd Pawlicki, Peter B. Dunscombe, Fundamentals and Clinical Applications
Arno J. Mundt, and Pierre Scalliet, Editors Eduardo Moros, Editor
ISBN: 978-1-4398-0436-0 ISBN: 978-1-4398-4890-6

Adaptive Radiation Therapy Emerging Imaging Technologies in


X. Allen Li, Editor Medicine
ISBN: 978-1-4398-1634-9 Mark A. Anastasio and Patrick La Riviere, Editors
ISBN: 978-1-4398-8041-8
Quantitative MRI in Cancer
Cancer Nanotechnology: Principles and
Thomas E. Yankeelov, David R. Pickens,
Applications in Radiation Oncology
and Ronald R. Price, Editors
Sang Hyun Cho and Sunil Krishnan, Editors
ISBN: 978-1-4398-2057-5
ISBN: 978-1-4398-7875-0
Informatics in Medical Imaging Monte Carlo Techniques in Radiation
George C. Kagadis and Steve G. Langer, Editors Therapy
ISBN: 978-1-4398-3124-3 Joao Seco and Frank Verhaegen, Editors
Adaptive Motion Compensation in ISBN: 978-1-4665-0792-0
Radiotherapy Image Processing in Radiation Therapy
Martin J. Murphy, Editor Kristy Kay Brock, Editor
ISBN: 978-1-4398-2193-0 ISBN: 978-1-4398-3017-8

Image-Guided Radiation Therapy Informatics in Radiation Oncology


Daniel J. Bourland, Editor George Starkschall and R. Alfredo C. Siochi,
ISBN: 978-1-4398-0273-1 Editors
ISBN: 978-1-4398-2582-2
Targeted Molecular Imaging
Michael J. Welch and William C. Eckelman, Cone Beam Computed Tomography
Editors Chris C. Shaw, Editor
ISBN: 978-1-4398-4626-1
ISBN: 978-1-4398-4195-0
Tomosynthesis Imaging
Proton and Carbon Ion Therapy
Ingrid Reiser and Stephen Glick, Editors
C.-M. Charlie Ma and Tony Lomax, Editors
ISBN: 978-1-4398-7870-5
ISBN: 978-1-4398-1607-3
Stereotactic Radiosurgery and
Comprehensive Brachytherapy: Stereotactic Body Radiation Therapy
Physical and Clinical Aspects Stanley H. Benedict, David J. Schlesinger, Steven
Jack Venselaar, Dimos Baltas, Peter J. Hoskin, J. Goetsch, and Brian D. Kavanagh, Editors
and Ali Soleimani-Meigooni, Editors ISBN: 978-1-4398-4197-6
ISBN: 978-1-4398-4498-4
Computer-Aided Detection and Diagnosis
Physics of Mammographic Imaging in Medical Imaging
Mia K. Markey, Editor Qiang Li and Robert M. Nishikawa, Editors
ISBN: 978-1-4398-7544-5 ISBN: 978-1-4398-7176-8
IMAGING IN MEDICAL DIAGNOSIS AND THERAPY
Series Editors: Andrew Karellas and Bruce R. Thomadsen

Published titles
Ultrasound Imaging and Therapy Handbook of Small Animal Imaging:
Aaron Fenster and James C. Lacefield, Preclinical Imaging, Therapy, and
Editors Applications
ISBN: 978-1-4398-6628-3 George Kagadis, Nancy L. Ford, Dimitrios N.
Karnabatidis, and George K. Loudos Editors
Cardiovascular and Neurovascular ISBN: 978-1-4665-5568-6
Imaging: Physics and Technology
Carlo Cavedon and Stephen Rudin, Editors Handbook of Radioembolization:
ISBN: 978-1-4398-9056-1
Physics, Biology, Nuclear Medicine,
and Imaging
Scintillation Dosimetry Alexander S. Pasciak, PhD., Yong Bradley, MD.,
Sam Beddar and Luc Beaulieu, Editors J. Mark McKinney, MD., Editors
ISBN: 978-1-4822-0899-3 ISBN: 978-1-4987-4201-6

Forthcoming titles

Physics of PET and SPECT Imaging


Magnus Dahlbom, Editor

Hybrid Imaging in Cardiovascular


Medicine
Yi-Hwa Liu and Albert Sinusas, Editors
Handbook of
Radioembolization
Physics, Biology, Nuclear Medicine, and Imaging

Edited by
Alexander S. Pasciak, PhD
J. Mark McKinney, MD
Yong C. Bradley, MD
CRC Press
Taylor & Francis Group
6000 Broken Sound Parkway NW, Suite 300
Boca Raton, FL 33487-2742

© 2017 by Taylor & Francis Group, LLC


CRC Press is an imprint of Taylor & Francis Group, an Informa business

No claim to original U.S. Government works

Printed on acid-free paper


Version Date: 20160808

International Standard Book Number-13: 978-1-4987-4201-6 (Hardback)

This book contains information obtained from authentic and highly regarded sources. Reasonable efforts have been made to
publish reliable data and information, but the author and publisher cannot assume responsibility for the validity of all materials
or the consequences of their use. The authors and publishers have attempted to trace the copyright holders of all material repro-
duced in this publication and apologize to copyright holders if permission to publish in this form has not been obtained. If any
copyright material has not been acknowledged please write and let us know so we may rectify in any future reprint.

Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmitted, or utilized in any
form by any electronic, mechanical, or other means, now known or hereafter invented, including photocopying, microfilming,
and recording, or in any information storage or retrieval system, without written permission from the publishers.

For permission to photocopy or use material electronically from this work, please access www.copyright.com (http://www.copy-
right.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923, 978-750-8400.
CCC is a not-for-profit organization that provides licenses and registration for a variety of users. For organizations that have been
granted a photocopy license by the CCC, a separate system of payment has been arranged.

Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used only for identifica-
tion and explanation without intent to infringe.

Library of Congress Cataloging‑in‑Publication Data

Names: Pasciak, Alexander S., editor. | McKinney, J. Mark, editor. | Bradley,


Yong C., editor.
Title: Handbook of radioembolization : physics, biology, nuclear medicine,
and imaging / edited by Alexander S. Pasciak, J. Mark McKinney, and Yong
C. Bradley.
Other titles: Imaging in medical diagnosis and therapy.
Description: Boca Raton : Taylor & Francis, 2017. | Series: Imaging in
medical diagnosis and therapy | Includes bibliographical references and
index.
Identifiers: LCCN 2016032428 | ISBN 9781498742016 (hardcover : alk. paper)
Subjects: | MESH: Liver Neoplasms--radiotherapy | Liver
Neoplasms--radionuclide imaging | Embolization, Therapeutic | Yttrium
Radioisotopes--therapeutic use | Tomography, Emission-Computed
Classification: LCC RC280.L5 | NLM WI 735 | DDC 616.99/4360757--dc23
LC record available at https://lccn.loc.gov/2016032428

Visit the Taylor & Francis Web site at


http://www.taylorandfrancis.com

and the CRC Press Web site at


http://www.crcpress.com
Dedication

To Alina…
Contents

Series preface xi
Preface xiii
Editors xv
Contributors xvii

Part 1 INtrODUCtION 1

1 Introduction to hepatic radioembolization 3


Andor F. Van Den Hoven, Daniel Y. Sze, and Marnix G.E.H. Lam

Part 2 PatIENt SELECtION aND trEatMENt PLaNNING 31

2 Treatment options for patients with primary and secondary liver cancer: An overview
of invasive, minimally invasive, and noninvasive techniques 33
Ricardo Paz-Fumagalli, David M. Sella, and Gregory T. Frey
3 Treatment planning part I: Vascular considerations associated with safety and efficacy
in radioembolization 53
Ray Bradford and J. Mark McKinney
4 Treatment planning part II: Procedure simulation and prognostication 65
Shyam M. Srinivas, Sankaran Shrikanthan, Naichang Yu, Susan D. Kost, Ram Gurajala,
and Karunakaravel Karuppasamy
5 Treatment planning part III: Dosimetric considerations in radioembolization with glass
and resin microspheres 87
Alexander S. Pasciak, Austin C. Bourgeois, and Yong C. Bradley
6 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy 113
Edward Kim, Joseph Titano, and Safet Lekperic

Part 3 trEatING PatIENtS WItH raDIOEMBOLIZatION 133

7 Radiation safety concerns associated with preparing the dosage, treating


and releasing the patient, and managing radioactive waste 135
William D. Erwin
8 The radiation biology of radioembolization 167
Marta Cremonesi, Francesca Botta, Mahila Ferrari, Lidia Strigari, Guido Bonomo,
Franco Orsi, and Roberto Orecchia

ix
x Contents

9 Microsphere deposition, dosimetry, radiobiology at the cell-scale,


and predicted hepatic toxicity 199
Stephan Walrand

Part 4 FOLLOWING PatIENtS trEatED WItH raDIOEMBOLIZatION 219

10 Postradioembolization imaging using bremsstrahlung 90Y SPECT/CT 221


S. Cheenu Kappadath
11 Quantitative postradioembolization imaging using PET/CT 229
Marco D’arienzo, Luca Filippi, and Oreste Bagni
12 Image-based three-dimensional dosimetry following radioembolization 251
Alexander S. Pasciak and S. Cheenu Kappadath
13 Diagnostic reporting using postradioembolization imaging 263
Yung Hsiang Kao
14 The use of postprocedural imaging in the medical management of patients 281
Austin C. Bourgeois, Marcelo S. Guimaraes, Yong C. Bradley,
Christopher Hannegan, and Alexander S. Pasciak

Part 5 NEW HOrIZONS 305

15 Future directions in radioembolization 307


Alexander S. Pasciak, J. Mark McKinney, and Yong C. Bradley

Index 323
Series preface

The advances in the science and technology of this challenge was the topic of conversation
medical imaging and radiation therapy are more during a lunch hosted by Taylor & Francis
profound and rapid than ever before since their Publishers and involving a group of senior medi-
inception over a century ago. Further, these dis- cal physicists (Arthur L. Boyer, Joseph O. Deasy,
ciplines are increasingly cross-linked as imaging C.-M. Charlie Ma, Todd A. Pawlicki, Ervin B.
methods become more widely used to plan, guide, Podgorsak, Elke Reitzel, Anthony B. Wolbarst,
monitor, and assess treatments in radiation therapy. and Ellen D. Yorke). The conclusion of this dis-
Today, the technologies of medical imaging and cussion was that a book series should be launched
radiation therapy are so complex and so computer- under the Taylor & Francis banner, with each vol-
driven that it is difficult for the persons (physicians ume in the series addressing a rapidly advancing
and technologists) responsible for their clinical use area of medical imaging or radiation therapy of
to know exactly what is happening at the point of importance to medical physicists. The aim would
care, when a patient is being examined or treated. be for each volume to provide medical physicists
The persons best equipped to understand the tech- with the information needed to understand tech-
nologies and their applications are medical physi- nologies driving a rapid advance and their appli-
cists, and these individuals are assuming greater cations to safe and effective delivery of patient
responsibilities in the clinical arena to ensure that care.
what is intended for the patient is actually delivered Each volume in the series is edited by one or
in a safe and effective manner. more individuals with recognized expertise in the
The growing responsibilities of medical physi- technological area encompassed by the book. The
cists in the clinical arenas of medical imaging and editors are responsible for selecting the authors of
radiation therapy are not without their challenges, individual chapters and ensuring that the chap-
however. Most medical physicists are knowledge- ters are comprehensive and intelligible to someone
able in either radiation therapy or medical imag- without such expertise. The enthusiasm of volume
ing and expert in one or a small number of areas editors and chapter authors has been gratifying
within their discipline. They sustain their exper- and reinforces the conclusion of the Minneapolis
tise in these areas by reading scientific articles and luncheon that this series of books addresses a
attending scientific talks at meetings. In contrast, major need of medical physicists.
their responsibilities increasingly extend beyond Imaging in Medical Diagnosis and Therapy
their specific areas of expertise. To meet these would not have been possible without the encour-
responsibilities, medical physicists periodically agement and support of the series manager, Luna
must refresh their knowledge of advances in medi- Han, of Taylor & Francis Publishers. The editors
cal imaging or radiation therapy, and they must be and authors, and most of all I, are indebted to her
prepared to function at the intersection of these steady guidance of the entire project.
two fields. How to accomplish these objectives is
a challenge. William Hendee
At the 2007 annual meeting of the American Asso- Founding Series Editor
ciation of Physicists in Medicine in Minneapolis, Rochester, Minnesota
xi
Preface

Primary and metastatic liver cancers are the fifth Radioembolization is a brachytherapy treatment
most commonly diagnosed cancer and the second delivered as part of a minimally invasive fluoro-
most common cause of cancer-related mortality in scopically guided intervention. In this procedure,
men worldwide, with slightly lower rates in women. millions of microscopic embolic spheres contain-
Environmental factors based on geographic loca- ing calibrated activities of either yttrium-90 (90Y)
tion carry a profound impact in primary liver can- or holmium-166 (166Ho) are infused into the right
cers. In some developing countries, liver cancer is or left hepatic artery where they embolize both
the most common form of cancer with higher rates tumor tissue and, to some extent, normal liver
of incidence secondary to viral hepatitis or expo- tissue. Because the hepatic artery primarily per-
sure to aflatoxin B1. Unfortunately, liver cancer fuses the tumor, greater concentrations of radio-
often carries a poor prognosis as surgical options active microspheres are trapped in the tumor
are commonly limited by multifocality combined compared to the normal liver. The relative differ-
with other factors, such as underlying liver disease. ence in microsphere concentration in tumor com-
External beam radiation therapy (EBRT) is pared to normal liver tissue following a successful
often a preferred treatment modality for many sur- radioembolization therapy can range anywhere
gically unresectable cancers, especially in concert from a factor of 2 to a factor of 15, providing the
with chemotherapy. However, for many years, its potential for sparing of healthy liver tissue com-
utility in the treatment of primary and secondary pared to conventional EBRT. However, there are
liver cancer has been limited by the high radio- other advantages. While a 40 Gy absorbed dose to
sensitivity of normal liver tissue. As is discussed normal liver tissue from EBRT could potentially
in the chapters of this book, 30–40 Gy represents cause liver failure, it is well below the toxic thresh-
a maximum tolerable dose to normal liver from old for a single-session treatment using radio-
EBRT, above which radiation-induced liver disease embolization, which is greater than 80 Gy. This
and liver failure are potentially fatal complica- unique paradox is due to differences in irradiated
tions. This fact has completely eliminated the use tumor volume, dose rate, and other factors, such as
of conventional whole-liver EBRT as a potential the heterogeneous microscopic dose distribution
treatment option for liver cancer. Newer advance- that results from radioembolization. This micro-
ments in image-guided intensity modulated radia- scopic absorbed dose heterogeneity, combined
tion therapy are greatly improving the prospects of with the regenerative propensity of healthy liver
EBRT as a viable treatment modality; however, the tissue, vastly reduces the toxicity of radioemboli-
high radiosensitivity of normal liver tissue, wors- zation and is a hallmark of the technique’s utility.
ened by underlying liver disease in many patients, Given the clear inherent benefits of the radio-
remains a challenge. embolization treatment, its use as a treatment
Despite technological advancements in EBRT, option for primary and metastatic liver cancer is
the most common form of radiation therapy for advancing extremely rapidly. While the methodol-
the treatment of primary and metastatic liver ogy behind radioembolization has been relatively
cancer is radioembolization, sometimes referred stable over the past 10 years, it is our belief that
to as selective internal radiation therapy or SIRT. this treatment is on the cusp of some rapid changes
xiii
xiv Preface

that will increase both its efficacy and the breadth with systemic chemotherapy, radio frequency,
of its clinical use. Our prediction is based on the cryo- and IRE percutaneous ablative tech-
following: niques with promising preliminary results.

● Commercial manufacturers of 90Y radioembo- While this is by no means an exhaustive list, it is


lization products are rapidly seeking approval still highly suggestive that the field of radioemboli-
for new hepatic treatment indications both in zation is poised for rapid advancement in the near
the United States and worldwide. Over the next future. Although generally considered a third- or
several years, on-label indications may match fourth-line palliative therapy for some forms of
what many leading institutions are currently metastatic liver cancer, some leading institutions
performing regularly as off-label treatment have moved radioembolization to a second-line
with radioembolization. treatment in combination with chemotherapy by
● New techniques in posttreatment quantitative taking advantage of new information and treat-
imaging will vastly expand the field’s under- ment planning techniques.
standing of the dose–response relationships Many of the recent advancements in radioem-
associated with radioembolization. These bolization are related more to radiation biology,
include the following: 90Y positron emission nuclear medicine, and the physics of the treatment
tomography/computed tomography (PET/CT), rather than the vascular aspects. As such, a book
quantitative bremsstrahlung single-photon focusing on these topics is appropriate, especially
emission computed tomography (SPECT)/CT, in light of the expected near-term growth and
and increasing use of directly imageable 166Ho advancement of the field. We expect that with
microspheres. Worldwide clinical trials are the expanded use of radioembolization, many
currently being planned to collect the data nec- individuals who have little prior experience with
essary to determine these dose–response rela- the procedure may pick up this book. While they
tionships using advanced imaging techniques. come from different backgrounds—medical physi-
● Alternatives to 90Y radioembolization, such cists, radiation oncologists, nuclear medicine radi-
as 166Ho radioembolization, are currently ologists, interventional radiologists, and health
under clinical use in some parts of the world. physicists—all have a necessary role to play in the
Alternative isotopes such as this provide execution of a well-planned radioembolization
certain advantages over 90Y, including effec- therapy. We suggest that regardless of background,
tive imaging with magnetic resonance imaging all individuals begin with Chapter 1, which
(MRI) and SPECT, and will undoubtedly lead expertly summarizes all aspects of the procedure
to expansion of the field of radioembolization in its entirety. The remaining chapters in this book
in the near future. fill in the details of radioembolization treatments
● Extrahepatic usage of radioembolization is as a currently valuable therapeutic method with
currently being investigated in clinical trials, many clinically relevant examples as well as some
including use for treatment of primary renal ideas that may aid the advancement of the field.
cell carcinoma.
● Multiple clinical trials are underway to assess Alexander S. Pasciak, J. Mark McKinney,
the utility of adjuvant 90Y radioembolization and Yong C. Bradley
Editors

Alexander S. Pasciak, PhD, earned a BS in electri- mentored residents, made numerous presenta-
cal engineering at the University of Washington, tions, and authored more than 60 peer-reviewed
MS in health physics and PhD in nuclear engineer- articles, abstracts, and book chapters. From 2008
ing at Texas A&M University. In 2010, Dr. Pasciak to 2012 Dr. McKinney was Chair of Radiology
completed a 2-year diagnostic medical physics at the University of Tennessee Medical Center
residency at the University of Texas MD Anderson and initiated the University of Tennessee inter-
Cancer Center after which he worked for 5 years ventional oncology radioembolization program.
as a diagnostic medical physicist at the University Dr. McKinney returned to Mayo Clinic in 2012 and
of Tennessee Medical Center in Knoxville, is serving as Chair of Radiology. Dr. McKinney is
Tennessee. Dr. Pasciak maintains a position as an an associate professor of radiology in the Mayo
associate professor of radiology at the University Clinic College of Medicine. Dr. McKinney is a
of Tennessee and is concurrently pursuing his MD recent past president of the Association of Program
degree at the Johns Hopkins University School of Directors in Radiology and is involved in the new
Medicine in Baltimore, Maryland. Dr. Pasciak is interventional radiology residency program.
active in multiple research endeavors in the fields
of interventional radiology and medical physics, Yong C. Bradley, MD, is an ABNM and ABR-
and he has published papers in high impact jour- certified nuclear medicine subspecialist at the
nals. Dr. Pasciak has published 35 articles in peer- University of Tennessee Medical Center in
reviewed medical journals, presented 62 research Knoxville, Tennessee. He completed his residency
abstracts at national meetings, has written 6 book training in radiology at Tripler Army Medical
chapters, and has filed 2 patents. He currently Center in Honolulu, Hawaii, and subsequently
serves as principal investigator on three externally completed a 2-year fellowship in nuclear medicine
funded research grants involving radioemboli- at Brooke Army Medical Center in San Antonio,
zation. Dr. Pasciak is certified by the American Texas. Dr. Bradley is a former chief of radiol-
Board of Radiology (ABR) in diagnostic medical ogy and nuclear medicine and molecular imag-
physics and is Mammography Quality Standards ing at the Brooke Army Medical Center, Wilford
Act (MQSA) qualified. Hall Medical Center, and San Antonio Military
Medical Center. Dr. Bradley has been interested
J. Mark McKinney, MD, earned a medical degree in cancer imaging and therapy for over 20 years.
at Loma Linda University School of Medicine in He has been involved in positron emission tomog-
California where he completed a diagnostic radiol- raphy (PET) and PET/computed tomography
ogy residency and interventional radiology fellow- (CT) imaging since 1998, when he was first intro-
ship. Dr. McKinney joined the Mayo Clinic in 1993. duced to PET along with radioimmunotherapy.
While at Mayo Clinic in Jacksonville, Florida he Presently, his interest has been concentrated in
developed the interventional radiology fellowship, liver radioembolization.

xv
Contributors

Oreste Bagni Gregory t. Frey


National Institute of Ionizing Radiation Metrology Mayo Clinic
Rome, Italy Jacksonville, Florida
Guido Bonomo Marcelo S. Guimaraes
European Institute of Oncology (IEO) Medical University of South Carolina
Milan, Italy Charleston, South Carolina
Francesca Botta ram Gurajala
European Institute of Oncology (IEO) Cleveland Clinic
Milan, Italy Cleveland, Ohio
austin C. Bourgeois
Christopher Hannegan
University of Tennessee Graduate School
Medical University of South Carolina
of Medicine
Charleston, South Carolina
Knoxville, Tennessee
ray Bradford Yung Hsiang Kao
Mayo Clinic Royal Melbourne Hospital
Jacksonville, Florida Melbourne, Australia

Yong C. Bradley S. Cheenu Kappadath


University of Tennessee Graduate School University of Texas MD Anderson Cancer Center
of Medicine Houston, Texas
Knoxville, Tennessee Karunakaravel Karuppasamy
Marta Cremonesi Cleveland Clinic
European Institute of Oncology (IEO) Cleveland, Ohio
Milan, Italy
Edward Kim
Marco D’arienzo Mount Sinai School of Medicine
National Institute of Ionizing Radiation Metrology New York, New York
Rome, Italy
Susan D. Kost
William D. Erwin
Cleveland Clinic
University of Texas MD Anderson Cancer Center
Cleveland, Ohio
Houston, Texas
Marnix G.E.H. Lam
Mahila Ferrari
University Medical Center
European Institute of Oncology (IEO)
Utrecht, The Netherlands
Milan, Italy
Luca Filippi Safet Lekperic
Santa Maria Goretti Hospital Mount Sinai School of Medicine
Latina, Italy New York, New York

xvii
xviii Contributors

J. Mark McKinney Shyam M. Srinivas


Mayo Clinic Cleveland Clinic
Jacksonville, Florida Cleveland, Ohio
roberto Orecchia Lidia Strigari
European Institute of Oncology (IEO) Regina Elena National Cancer Institute (IFO)
Milan, Italy Rome, Italy
Franco Orsi Daniel Y. Sze
European Institute of Oncology (IEO) Stanford University Medical Center
Milan, Italy Stanford, California
alexander S. Pasciak Joseph titano
University of Tennessee Graduate School Mount Sinai School of Medicine
of Medicine New York, New York
Knoxville, Tennessee andor F. Van Den Hoven
ricardo Paz-Fumagalli University Medical Center
Mayo Clinic Utrecht, The Netherlands
Jacksonville, Florida Stephan Walrand
David M. Sella Catholic University of Louvain
Mayo Clinic Louvain, Belgium
Jacksonville, Florida Naichang Yu
Sankaran Shrikanthan Cleveland Clinic
Cleveland Clinic Cleveland, Ohio
Cleveland, Ohio
PART 1
Introduction

1 Introduction to hepatic radioembolization 3


Andor F. Van Den Hoven, Daniel Y. Sze, and Marnix G.E.H. Lam
1
Introduction to hepatic
radioembolization

ANDOR F. VAN DEN HOVEN, DANIEL Y. SZE, AND MARNIX G.E.H. LAM

1.1 General introduction 3 1.4 Treatment 17


1.1.1 What is radioembolization? 3 1.4.1 Medication and periprocedural
1.1.2 A brief history of radioembolization 4 care 17
1.1.3 Indications for radioembolization 6 1.4.2 Treatment technique 17
1.1.4 Comparison of radioembolization 1.4.3 Catheter types and particle-
and external beam radiation fluid dynamics 18
therapy principles 7 1.4.4 Imaging of the therapeutic
1.2 Types of microspheres and microsphere distribution 18
radionuclides used 8 1.4.5 Dose–response relationship 19
1.2.1 Yttrium-90 microspheres 8 1.5 Treatment-related laboratory and
1.2.2 Holmium-166 microspheres 10 clinical toxicity 19
1.3 Pretreatment workup 11 1.5.1 Complaints during treatment 19
1.3.1 Laboratory and clinical 1.5.2 Laboratory toxicity 19
investigations 11 1.6 Tumor response assessment 21
1.3.2 Pretreatment imaging: liver 1.6.1 Anatomical tumor response
CT/MRI and 18F-FDG-PET 11 assessment 22
1.3.3 Assessing the individual 1.6.2 Functional tumor response
hepatic arterial anatomy 12 assessment 22
1.3.4 Preparatory angiography and 1.7 Conclusion 23
intraprocedural imaging 13 References 23
1.3.5 Imaging of the scout dose
distribution 15
1.3.6 Pretreatment activity calculations 16

vasculature to irradiate liver tumors from within.


1.1 GENEraL INtrODUCtION This therapy is based on the principle that liver
tumors are almost exclusively vascularized by the
1.1.1 WHAT IS RADIOEMBOLIZATION? hepatic artery, whereas the healthy liver tissue
receives the majority of its blood supply from the
Radioembolization is a therapy during which portal vein. Therefore, following the administra-
radioactive microspheres are administered through tion in the hepatic artery, microspheres will be
a microcatheter placed in the hepatic arterial carried preferentially toward the distal arterioles
3
4 Introduction to hepatic radioembolization

in and around tumors. Clusters of microspheres latest introductions are radiation segmentectomy
are formed inside and in the periphery of tumors, as a potentially curative technique to eradicate
where they emit high-energy β-radiation to induce focal solitary liver tumors (Riaz et al., 2011), down-
cell death, while relatively sparing the healthy liver staging of unresectable disease to enable poten-
tissue (Braat et al., 2015). Radioembolization is a tially curative surgical resection or transplantation
minimally invasive, image-guided, locoregional (Braat et al., 2014), and radiation lobectomy to
alternative, or adjunct to more conventional thera- induce contralateral hypertrophy as an alternative
pies such as surgery, systemic chemotherapy, and to portal vein embolization in surgical candidates
external beam radiation therapy for patients with (Gaba et al., 2009; Vouche et al., 2013). Additional
liver-dominant malignancy. The advantages of this information on these techniques is presented in
treatment are the targeted delivery of a very high Chapter 6. Applying radioembolization principles
radiation-absorbed dose to tumors, with limited to the treatment of solid tumors in organs other
systemic side effects and hepatotoxicity (Kennedy, than the liver has also been provisionally explored,
2014). but falls outside the scope of this book.
The efficacy and safety of radioembolization
have been proven in patients with primary liver 1.1.2 A BRIEF HISTORY OF
tumors such as hepatocellular carcinoma (HCC) RADIOEMBOLIZATION
(Hilgard et al., 2010) and intrahepatic cholangio-
carcinoma (ICC) (Mouli et al., 2013), as well as Several earlier studies and discoveries have set the
in metastatic liver tumors from various primary backdrop for the clinical development of radio-
tumors, with colorectal cancer (CRC) (Kennedy embolization as a technique to treat liver tumors.
et al., 2015), breast cancer (BrC), neuroendocrine These investigations showed that large quanti-
tumors (NET) (Devcic et al., 2014), and uveal ties of glass microspheres could be safely admin-
melanoma (Xing et al., 2014) being the most com- istered intra-arterially in animal experiments
mon. Typically, radioembolization is performed as (Prinzmetal and Ornitz, 1948), that radioactive
a stand-alone treatment in salvage patients with gold-covered charcoal particles administered
liver-dominant disease, but several clinical trials intravenously or yttrium oxide particles admin-
are currently evaluating its role in earlier lines of istered via a pulmonary artery catheter could be
treatment and in combination with systemic ther- used to treat lung cancer patients successfully
apy or other locoregional treatments such as radio- (Muller and Rossier, 1951), and that liver tumors,
frequency ablation. even ones that reached the liver via the portal cir-
“Radioembolization” is used as an umbrella culation, were preferentially vascularized by the
term for the treatment of liver tumors with hepatic artery when they exceeded about 50 μm
varying disease extents ranging from a single in diameter (Bierman et al., 1951). The first report
focal subsegmental liver tumor to extensive dis- on radioembolization was published in 1960 by
seminated or infiltrative disease, which can be the American surgeon Edgar D. Grady and his
hypo- to hypervascular in nature, situated in liv- colleagues, affiliated with Piedmont Hospital and
ers that are relatively healthy, cirrhotic, partially Georgia Institute of Technology in Atlanta, GA,
resected, transplanted, or heavily pretreated with USA (Grady et al., 1960). Subsequent preclinical
systemic or intra-arterial chemotherapy. These and clinical investigations by Kim et al. (1962),
situations pose various challenges and require Caldarola et al. (1964), Blanchard et al. (1965a),
other approaches with regard to safety precau- and Ariel (1965) followed shortly thereafter.
tions, treatment planning and dose calculation, However, technical aspects such as the method to
microsphere type usage, and catheter positioning access the hepatic vasculature, the site of admin-
during administration. Furthermore, treatment istration, safety precautions, size and material of
techniques and strategies are dependent on oper- the particles, and the radioactive isotope and the
ator experience and preferences and may differ amount of activity to be infused still needed to be
considerably among practices. refined in the years to follow.
Research continues to provide new insights into Experiments with New Zealand rabbits demon-
how to optimize radioembolization treatment, strated that injection of radioactive microspheres
and new indications continue to arise. Among the via the hepatic artery established preferential tumor
1.1 General introduction / 1.1.2 A brief history of radioembolization 5

targeting, whereas injection via the portal vein did Determining the optimal treatment activity
not (Blanchard et al., 1965a), which echoed early (pretreatment dosimetry) has been a challenge
clinical results in humans (Grady, 1979). However, from the start (Blanchard et al., 1965b). It was
it proved challenging to catheterize the hepatic already recognized that the intrahepatic micro-
artery in both animals and humans. Access sphere distribution is highly heterogeneous after
methods included antegrade catheterization of the treatment, but imaging methods available at that
celiac artery via brachial artery access, retrograde time precluded the assessment of the tissue mass
catheterization through femoral arteriotomy with exposed to radiation. Therefore, treatment activity
the use of a balloon below the level of the celiac could not be adapted to effective tumor-absorbed
artery, and catheterization of the hepatic artery dose and safe healthy liver-absorbed dose values.
by accessing the gastroepiploic artery during Instead, the required treatment activity was calcu-
laparotomy. lated based on a target whole liver-absorbed dose
After trial and error it was learned that addi- of 5000 rad (50 Gy), which had been demonstrated
tional safety precautions were required, since as a safe dose in animal experiments. Doses were
extrahepatic deposition of radioactive micro- prescribed based on the formula that per gram
spheres (in the gastrointestinal tract or lungs) as of liver tissue 1 mCi (37 MBq) would be required
well as too much radiation exposure of the healthy to deliver an absorbed dose of 182 rad (1.82 Gy)
liver tissue could result in life-threatening com- (Grady, 1979).
plications (Blanchard et al., 1965b). Therefore, The first efficacy reports were case series
routine “skeletonization” (a surgical term used to reporting posttreatment survival and the clinical
describe isolation of the main vascular trunk by condition of patients with primary or metastatic
ligating all side branches) of the hepatic artery, liver cancer. These results were generally promis-
as well as injection and imaging of radiolabelled ing, and some cases showed unprecedented dis-
albumin particles before treatment to simulate the ease control, but these reports were written prior
therapeutic microsphere distribution, was advo- to the availability of computed tomography, mag-
cated and eventually became standard of practice netic resonance imaging, and quantitative ultra-
(Grady, 1979; Ariel and Padula, 1982). sonography. Patients with inoperable disease had
Initially, glass microspheres of 50–100 µm diam- no good alternatives at that time, since the effec-
eter were used. Soon, however, it was recognized tiveness of systemic chemotherapy and external
that smaller resin microspheres (15–30 µm) were beam radiation therapy remained disappointing.
easier to keep in suspension and would still not In 1989, Gray et al. published the first prospec-
pass through the capillaries. After several years tive trial results on radioembolization demon-
of experimentation with other isotopes such as strating an objective treatment response, defined
Phosphorus-32 (32P) (Caldarola et al., 1964; Grady as a decline of carcinoembryonic antigen (CEA)
et al., 1975), Yttrium-90 (90Y) established its domi- levels after treatment in 9/10 treated patients with
nance. Reported benefits of 90Y included a pure colorectal cancer liver metastases (Gray et al.,
high-energy yield of tumoricidal β-radiation (max 1989). In the next two decades, only a few prospec-
energy of 2.28 MeV), a short soft-tissue penetra- tive studies followed patients with primary liver
tion (max 11 mm), and a 64-h half-life, which lim- cancer and colorectal liver metastases (Lau et al.,
ited potential safety hazards for persons in close 1994; Rosler et al., 1994; Gray et al., 2001). Among
proximity to a treated patient. Early reports did, these studies was the first randomized controlled
however, acknowledge the importance of imag- trial, which demonstrated that the addition of
ing the posttreatment microsphere distribution radioembolization to regional hepatic arterial
and the limited possibilities inherent to the use of chemotherapy (floxuridine) in salvage patients
90
Y (Grady et al., 1963; Ariel, 1965). The secondary with colorectal cancer liver metastases resulted in
bremsstrahlung γ-ray produced by β-activity significantly improved tumor response.
could be detected with a Geiger–Muller survey Eventually, 90Y-microspheres received Conformité
meter or a scintillation crystal probe. Ariel even Européenne (CE) mark in the European Union and
added Ytterbium-169 (169Yb; γ-ray 52–310 keV; T1/2 U.S. Food and Drug Administration (FDA) approval
32 days) to the microspheres as a radiation source in the United States for the treatment of HCC and
for imaging with a γ-camera (Ariel, 1965). metastatic colorectal cancer, which in turn led to a
6 Introduction to hepatic radioembolization

broader availability of radioembolization to patients controlled trials. These topics will be discussed in
and a renewed scientific interest. more detail in Chapter 15.
The past two decades have been characterized
by an enormous growth in the widespread use
of radioembolization to treat salvage patients, 1.1.3 INDICATIONS FOR
with either primary or metastatic liver cancer. RADIOEMBOLIZATION
It is increasingly acknowledged that, as long
as the liver disease is the survival-limiting fac- At this moment, the indication for radioemboli-
tor in the patients’ prognosis, radioembolization zation as a stand-alone therapy for patients with
treatment is expected to be beneficial in patients liver metastases is primarily based on unresect-
with all kinds of liver-dominant tumor types. able, liver-dominant metastases refractory to
Patient selection, workup, treatment technique, standard systemic therapy. The standard for sys-
and analyses of treatment toxicity and response temic therapy differs per primary tumor type and
have all been vastly improved. Modern imag- per geographical location, and may include cyto-
ing techniques including multidetector contrast- toxic chemotherapeutic agents as well as targeted
enhanced computed tomography (CT), magnetic small molecules, monoclonal antibodies, and
resonance imaging (MRI), and C-arm cone beam immunomodulators. The prevailing principle is
CT now allow for a detailed assessment of tumor that no other therapy should be available with
location, tumor characteristics, and individual more convincing scientific evidence of effective-
hepatic arterial anatomy before treatment. This ness. Patients with contraindications to or unac-
enables the operator to set a feasible individual- ceptable toxicity from systemic therapy are also
ized treatment strategy with the aim to achieve eligible. Since large randomized controlled stud-
adequate tumor targeting, while minimizing the ies are currently investigating the role of radio-
chance of treatment-related complications. The embolization combined with systemic therapy in
advent of nuclear medicine imaging techniques the first- and second-line treatment of colorectal
such as single photon emission computed tomog- cancer liver metastases, radioembolization may
raphy (SPECT)/computed tomography (CT) and potentially be performed earlier in the treatment
positron emission tomography (PET)/CT, as cycle in the future.
well as the development of non-90Y microspheres In patients with HCC, radioembolization is
such as Holmium-166 (166Ho) microspheres, has generally reserved for patients with intermedi-
enabled imaging of the particle distribution and ate and early advanced disease stages (Braat et al.,
quantification of radiation-absorbed doses. It is 2015). These are patients with large multinodular
now possible to identify an unfavorable particle tumors (>3, ≥3 cm), with or without macrovascu-
distribution early on when the treatment plan can lar invasion, sufficient liver function (Child–Pugh
still be modified. Tumor response assessment is A–B), and an acceptable clinical condition [World
also becoming less observer dependent with all Health Organization (WHO) performance status
the possibilities that functional MRI and 18-flu- score 0–2], corresponding to Barcelona Clinic
oro-2-deoxyglucose positron emission tomogra- Liver Cancer staging system stages B–C (Forner
phy (18F-FDG-PET) imaging have to offer. et al., 2014). Some patients may have already failed
The challenges for the near future will be to chemoembolization and/or systemic treatment
clarify which patients will benefit most from with sorafenib, but radioembolization is offered
radioembolization, to improve methods for as an alternative to chemoembolization in some
treatment activity calculations, to maximize practices, even for earlier stage disease.
treatment efficacy, to reduce treatment-related Treatment with radioembolization should be
toxicity, to standardize treatment technique, to considered relatively aggressive, and must be tech-
enhance our understanding of relevant particle- nically feasible and clinically tolerable. Additional
fluid dynamics, radiobiology, and systemic treat- important criteria for patient selection are summa-
ment effects, to explore combination therapies, rized in Table 1.1. It should be noted that indica-
and to strengthen scientific evidence by prov- tions and contraindications are subject to change
ing superiority over conventional and emerging over time as clinical experience, both positive and
therapies in large-scale phase III randomized negative, accumulate over the years.
1.1 General introduction / 1.1.4 Comparison of radioembolization and external beam radiation 7

Table 1.1 Eligibility criteria and contraindications for radioembolization

Contraindications
Eligibility criteria relative absolute
Good clinical condition WHO/ECOG PS >2a
Life expectancy ≥3 months Life expectancy <3
months
Adequate vital functions Mild laboratory abnormalities Severe laboratory
abnormalities
indicating critical liver,
renal, or bone-marrow
failure
Adequate portal venous liver Portal vein thrombosisb
vascularization Prior portal vein embolization or main
portal vein occlusion
Adequate functional hepatic Cirrhotic liver (Child–Pugh score >B7), Uncompensated liver
reserve total bilirubin >2.0 mg/dL (=34.2 failure
μmol/L)
Liver tumor burden <70% Liver tumor burden ≥70%
Undisturbed biliary system Biliary stent or prior sphincterotomy, Active cholangitis
choledochojejunostomy, or
hepaticojejunostomyc
Adequate arterial access Celiac axis and superior mesenteric Occluded intrahepatic
artery occluded arterial network
Successful preparatory Uncorrectable
angiographic procedure gastrointestinal
microsphere
deposition; expected
lung dose >30 Gy (or
50 Gy cumulative)
Interval since last dose of Interval since last dose of systemic Active use of
systemic therapy ≥4 weeks therapy <4 weeks antiangiogenic agents
(bevacizumab,
aflibercept)
Note: The most important eligibility criteria and associated contraindications for radioembolization are presented
in this table. A distinction between relative and absolute contraindications is made. ECOG PS, World Health
Organization/Eastern Cooperative Oncology Group Performance Score.
a
Corresponds to Karnofsky score <50.
b
No contraindication in HCC.
c
May require antibiotic prophylaxis to prevent liver abscesses.

1.1.4 COMPARISON OF from external beam radiation therapy (EBRT).


RADIOEMBOLIZATION AND Both therapies aim to achieve tumor cell death
EXTERNAL BEAM RADIATION through radiation-induced apoptosis and pro-
THERAPY PRINCIPLES liferative capacity inhibition (Eriksson and
Stigbrand, 2010). However, differences in the
Radioembolization is also referred to as selec- technical methods to target the tumor have an
tive internal radiation therapy (SIRT), indicat- important impact on the radiation distribu-
ing that it is a preferentially tumor targeted tion, as well as the dose rate and the possibility
form of brachytherapy that differs considerably to fractionate the treatment dose, which in turn
8 Introduction to hepatic radioembolization

determines inherent strengths and limitations of rates and liver toxicity, and that the accurate predic-
both treatment modalities. tion of the therapeutic microsphere distribution is a
Conventional whole-liver EBRT is no longer great challenge.
considered a viable treatment option. The healthy Additional detailed discussion on the hepatic
liver tissue is very sensitive to radiation, and whole- radiation biology of radioembolization and EBRT
liver-absorbed doses exceeding 30 Gy are associated are presented on macroscopic and microscopic lev-
with an increased risk of potentially fatal radiation- els in Chapters 8 and 9, respectively.
induced liver failure (Emami et al., 1991; Fuss et al.,
2004; Sharma, 2014). This phenomenon is even more
pronounced in patients with cirrhotic liver disease, 1.2 tYPES OF MICrOSPHErES
or a prior history of hepatotoxic systemic therapy. aND raDIONUCLIDES USED
However, the introduction of image-guided, confor-
mal, intensity modulated, stereotactic body radia-
tion therapy (SBRT) has made it possible to achieve Different types of radioactive microspheres are
highly preferential tumor targeting, thereby sparing commercially available. The type of microspheres
much of the adjacent and remote healthy liver tissue can be divided based on the embedded radioac-
(Fuss et al., 2004; Sharma, 2014). A safety margin of tive isotope (90Y or 166Ho) or microsphere material
up to 2 cm surrounding the tumor is required, which (resin, glass, or poly-l-lactic acid). These micro-
can result in some hepatotoxicity. Three-dimensional spheres all have different production processes,
images of the liver are acquired during a simulation physical characteristics, and methods of use. The
procedure, allowing drafting of a detailed therapy most important characteristics of the different
plan. Patients subsequently undergo multiple treat- microsphere types are summarized in Table 1.2.
ment sessions, during which photon beams pro-
duced by a linear accelerator apply small fractions 1.2.1 YTTRIUM-90 MICROSPHERES
of the total radiation dose. The greatest advantage of
90
EBRT is that the radiation can be actively targeted Y is a nearly pure (99.99%) β-emitter. It has a half-
with precision, yielding a predictable dose and treat- life of 64.1 h and decays to stable Zirconium-90
ment effect. The most important disadvantages are (90Zn). With maximum beta particle (β–) energy of
that it is not feasible to treat patients with a high 2.28 MeV—resulting in an energy release of 49.67
tumor burden, and the maximum radiation dose J/GBq—and a range in water or soft-tissue of 2.5
delivered is still limited by potential hepatotoxicity. mm (mean) and 11 mm maximum, it is a suitable
Radioembolization, on the contrary, depends on a radionuclide to treat cancer with an appropriate
hemodynamic mechanism for tumor targeting. The safety profile. Radioactive 90Y can either be pro-
interplay between catheter positioning, particle-fluid duced by neutron irradiation of stable Yttrium-89
dynamics, and tumor vascularization ultimately (89Y) or by chemical separation from the parent
determines the dose delivered to tumors and to nor- isotope Strontium-90 (90Sr), a fission product of
mal liver. Pathological examination of explanted liv- uranium (Walker, 1964). Imaging of the radiation
ers that were treated with radioembolization showed emission from 90Y is a challenge due to the absence
that the posttreatment microsphere distribution of γ-radiation emission. SPECT images can only be
was highly heterogeneous. Furthermore, the micro- acquired by the detection of bremsstrahlung, sec-
spheres tended to cluster preferentially in the periph- ondary γ-radiation produced by slowing of the beta
ery of the tumors with a concentration up to 200 particles in tissue (Figure 1.1a), a modality with very
times greater than in the tumor core or in healthy limited spatial resolution. Actually, 90Y has a minor
liver tissue (Campbell et al., 2001; Kennedy et al., branch to the first excited state of 90Zn at 1.76 MeV
2004). Therefore, the greatest benefits of radioem- (0+–0+ transition). As a result, once in every 32 mil-
bolization are that very high tumor-absorbed doses lion (31.86 × 106) decays, an electron–positron (β–/
can be achieved and that treatment feasibility is less β+) pair is created. This process is called internal-pair
dependent on tumor burden. However, inherent dis- production and enables positron emission detec-
advantages are that tumor targeting can turn out to tion with PET at high 90Y-activities (Figure 1.1a)
be suboptimal in patients with relatively hypovascu- (D’Arienzo, 2013). SPECT and PET imaging of 90Y
lar tumors, yielding highly variable tumor response are discussed further in Chapters 10 and 11.
1.2 Types of microspheres and radionuclides used / 1.2.1 Yttrium-90 microspheres 9

Table 1.2 Microsphere characteristics


Isotope Yttrium-90 (90Y) Holmium-166 (166Ho)
Half-life 64.1 h 26.8 h
Decay product Zirconium-90 (90Zn) Erbium-166 (166Er)
Radiation emission β (max 2.28 MeV) β (max 1.74 and 1.85),
γ (max 81 and 1.38 keV)
Energy per activity 49.67 J/GBq 15.87 J/GBq
Tissue penetration 2.5 mm mean, 11 mm maximum 2.5 mm mean, 8.4 mm maximum
Imaging PET (internal-pair production) SPECT (γ-imaging)
SPECT (bremsstrahlung) MRI (R2* mapping)
Material Glass Resin PLLA
(ceramic)
Product name TheraSphere® SIR-Spheres® QuiremSpheres®
Size 20–30 μm 32.5 ± 5 μm 20–50 μm
Density 3.3 g/cc 1.6 g/cc 1.4 g/cc
Spheres per vial 1.2–8 × 106 40–80 × 106 33 × 106
Specific activity per sphere 2500 Bq 40–70 Bq 450 Bq
Max activity per dose 20 GBq 3 GBq 15 GBq
Dosimetry method MIRD based BSA method MIRD based
recommended by
manufacturer
Embolic effect Low Moderate Moderate
Note: The characteristics of the glass and resin yttrium-90 microspheres and holmium-166 microspheres are sum-
marized in this table. PLLA, poly-l-lactic acid.

Glass and resin 90Y-microspheres currently have 10–20 times less particles need to be injected than
near worldwide commercial availability. In 2002, with resin microspheres to administer the same
resin 90Y-microspheres received FDA approval treatment activity. As a consequence, the embolic
for the treatment of metastatic colorectal cancer effect is much smaller during injection, so the entire
when given in conjunction with floxuridine, and treatment dose can be injected at once with a lower
received CE mark for the treatment of inoperable risk of stasis and particle reflux.
liver tumors. Glass 90Y-microspheres received FDA
approval under a humanitarian device exemption 1.2.1.2 Resin microspheres
in 1999 for the treatment of patients with HCC,
including those with branch portal vein thrombo- The production process of resin 90Y-microspheres
sis, and received CE mark in 2005 for the treatment (SIR-Spheres®, Sirtex Medical Limited, North
of hepatic neoplasms. Sydney, Australia) is different; in this type of
microsphere, 90Y cations in solution are chemi-
1.2.1.1 Glass microspheres cally incorporated onto the bland microsphere
surface by binding to the carboxylic group of the
Glass 90Y-microspheres (TheraSphere®, Nordion Inc. acrylic polymer matrix (Gulec and Siegel, 2007;
for BTG International, Ottawa, ON, Canada) are Giammarile et al., 2011). Resin microspheres have
produced by incorporating 89Y oxide into the glass a much lower density than glass microspheres,
matrix of the microsphere and subsequent activa- which could potentially result in a more distal dis-
tion by neutron bombardment in a nuclear reactor tribution in the tumor vasculature (Jernigan et al.,
facility (Wollner et al., 1988). Compared with the 2015). Furthermore, the relatively low specific
other microsphere types, glass 90Y-microspheres activity requires injection of a higher number of
have a relatively high density, and a high specific microspheres, approximately 20-80 million. Since
activity per sphere (2500 Bq/sphere). Therefore, this involves a greater embolic effect, stasis of blood
10 Introduction to hepatic radioembolization

Yttrium-90 Holmium-166

SPECT PET MRI SPECT


Bremsstrahlung γ511 keV, 32 positrons γ81 keV
x-rays per million decays
B0

Bremsstrahlung
e–
e– x-rays +

90Y e+ e– – 166Ho –

γ511 keV

(a) (b)

Figure 1.1 Schematic illustration of the physical properties and decay of 90Y (a) and 166Ho (b) in the
liver. The upper row displays examples of the microsphere biodistribution in the liver using 90Y brems-
strahlung SPECT (a, upper left), 90Y PET (a, upper right) 166Ho-MRI (b, upper left), and 166Ho-SPECT
(b, upper right). (With kind permission from Springer Science+Business Media. Cardiovasc Intervent
Radiol, Radioembolization dosimetry: The road ahead, 38, 2014, Smits et al.)

flow  may occur during administration. Therefore, dose in liver tissue (Prince et al., 2014a). The bio-
resin 90Y-microspheres must be administered care- distribution of 166Ho-microspheres can be visual-
fully by hand injection in smaller aliquots, with ized on SPECT (Figure 1.1b), using the low-energy
intervening angiography to reevaluate pace of flow γ-radiation (81 keV, 6.2%; 1.38 keV, 0.93%), and
and degree of stasis. Glass and resin microspheres with magnetic resonance imaging, utilizing the
may be used in different or similar tumor types paramagnetic properties of 166Ho (Figure 1.1b)
and disease extents, but it remains controversial (Smits et al., 2013a).
165
how the differences in distribution patterns impact Ho acetylacetonate poly(l-lactic acid) par-
treatment efficacy. ticles are manufactured through a solvent evapo-
ration process, and subsequently activated in a
1.2.2 HOLMIUM-166 nuclear reactor facility to create radioactive 166Ho
MICROSPHERES acetylacetonate poly(l-lactic acid) microspheres
(Nijsen et al., 2001; Zielhuis et al., 2006). The den-
The isotope 166Ho emits both high-energy sity, size, specific activity per sphere, number of
β-radiation and low-energy γ-radiation. It has injected 166Ho-microspheres, and embolic effect
a shorter half-life than 90Y (26.8 h) and decays are comparable with resin 90Y-microspheres.
166
with a relatively high dose rate to the stable ele- Ho-microspheres received CE mark approval,
ment erbium-166 (166Er). 166Ho emits β-radiation but no FDA approval yet. The transition toward
at two energy levels, maximum 1.74 MeV (48.7%) commercial availability in Europe is ongoing. The
and 1.85 MeV (50%), with a maximum soft-tissue safety of 166Ho-radioembolization has been dem-
range of 8.4 mm. The resulting energy release is onstrated in a phase I dose escalation study in
much lower (15.87 J/GBq) than with 90Y; there- patients with unresectable chemorefractory liver
fore, a larger administered treatment activity is metastases from various primary tumor types,
required to achieve the same radiation-absorbed and the results of a phase II clinical trial that
1.3 Pretreatment workup / 1.3.2 Pretreatment imaging: Liver CT/MRI and 18F-FDG-PET 11

investigated treatment efficacy are expected to be be recorded during clinical investigation. The
released soon. general clinical status can be summarized by per-
formance status (as defined by the WHO/Eastern
Cooperative Oncology Group): 0—asymptomatic,
1—symptomatic but completely ambulatory, 2—
1.3 PrEtrEatMENt WOrKUP symptomatic <50% of time in bed, 3—symptomatic
>50% of time in bed but not bedbound, 4—bed-
A thorough pretreatment workup is essential to bound, 5—dead. A performance status >2 is
screen patients for treatment eligibility, and to considered an exclusion criterion for radioemboli-
ensure that radioembolization treatment is per- zation. Occasionally, a poorer performance status
formed as safely and effectively as possible. A may reflect toxicity from other therapies that is
standard workup includes laboratory and clini- expected to improve after discontinuation of those
cal investigations, pretreatment cross-sectional therapies, a potential exception to the guidelines.
imaging, a preparatory angiographic procedure,
simulation scout dose imaging, and pretreatment 1.3.2 PRETREATMENT IMAGING:
activity calculation. LIVER CT/MRI AND 18F-FDG-PET
1.3.1 LABORATORY AND CLINICAL Cross-sectional pretreatment imaging is used for
INVESTIGATIONS the evaluation of the liver parenchyma, vascula-
ture, and presence and extent of extrahepatic dis-
Laboratory and clinical investigations are used to ease. CT, magnetic resonance imaging (MRI), and
assess vital functions and general clinical status, whole-body 18F-FDG-PET all play an important
and to record baseline values as a reference for tox- role.
icity assessments. For the evaluation of the liver parenchyma—
The laboratory investigations should include characterization and localization of liver tumors
parameters to assess hepatobiliary function (albu- and their relation with surrounding vessels and
min, total bilirubin, gamma-glutamyl transferase, biliary ducts—MRI is superior to CT in terms of
alkaline phosphatase, alanine aminotransferase, soft-tissue contrast. Dynamic contrast-enhanced
aspartate aminotransferase), renal function (creat- sequences can be used to assess tumor hypervas-
inine, estimated glomerular filtration ratio), coag- cularity and washout in a larger number of phases,
ulation status (platelet count, prothrombin time, while diffusion weighted and T2-weighted imaging
activated partial thromboplastin time, thrombin provide options for high-sensitivity tumor detec-
time, internationalized normalized ratio), and tion. Imaging with CT is faster, cheaper, higher
hematological function (hemoglobin and hemato- spatial resolution, and less susceptible to motion
crit, white blood cell count). Additional parameters artifacts, but this comes at the cost of ionizing
to consider are tumor markers (carcinoembryonic radiation and iodinated contrast agent burden.
antigen [CEA], alpha-fetoprotein [AFP], chromo- When using CT, multiphasic images (arterial, por-
granin-A [CgA], etc., depending on the tumor type tal venous, equilibrium phase) should be acquired
and biomarker secretion), and indicators for an and adequate timing of these phases is especially
acute infection (C-reactive protein) or cell tissue important to depict tumor types with different
damage (lactate dehydrogenase). degrees of arterial vascularization. A late arterial
Deviations from reference values are expected, phase and an equilibrium phase are recommended
considering the severity of disease in most patients. for the imaging of hypervascular tumors, while
However, severe deviations (Common Terminology relatively hypovascular tumors are often best visu-
Criteria for Adverse Events or CTCAE grades 3–4) alized on a portal venous phase (Rengo et al., 2011).
should be considered indicators of organ dysfunc- An arterial phase and a portal venous phase
tion, which may render radioembolization treat- are required for vascular assessment. The arterial
ment unsafe. phase can be used to evaluate accessibility of the
Previous treatments, chronic diseases, recent hepatic artery (through the celiac axis and superior
periods of acute illness, current medication use, mesenteric artery), to reveal the individual hepatic
and allergies (especially for contrast agents) should arterial anatomy including variants and parasitized
12 Introduction to hepatic radioembolization

arteries, and to identify tumor-feeding branches. vein can be used to indicate this division on cross-
The portal venous phase allows for evaluation of sectional imaging. The right hemi-liver is divided
the portal and hepatic veins for patency and tumor into an anterior and posterior sector by the right
invasion, and it confirms segmentation of the liver. portal scissura, as indicated by the course of the
The importance of oncological 18F-FDG-PET right hepatic vein. The level of the portal bifurcation
imaging is increasingly acknowledged. 18F-FDG- marks the distinction of superior and inferior seg-
PET has a high sensitivity for the detection of liver ments in these sectors (segments 5/8 anterior, and
metastases, especially in patients with tumor types segments 6/7 posterior). The left hemi-liver is simi-
that are expected to be PET avid (cholangiocarci- larly divided into an anterior and posterior sector
noma, liver metastases from CRC, BrC, and uveal by the left portal scissura, as indicated by the course
melanoma) (Tsurusaki et al., 2014). Furthermore, of the left hepatic vein. The fissure for the falciform
18
F-FDG-PET shows more extrahepatic lesions ligament divides segment 3 and 4 in the left anterior
than CT during radioembolization workup in sector, whereas segment 2 forms the only segment
patients with metastatic colorectal cancer (mCRC), of the left posterior sector. Segment 4 can also be
which can lead to considerable changes in man- divided into cranial and caudal subsegments 4a and
agement (Rosenbaum et al., 2013). In patients 4b. Segment 1 is situated between the portal bifur-
with HCC and NET liver metastases, the role of cation and the inferior vena cava. This independent
18
F-FDG-PET remains secondary, due to a less segment receives small branches from the left and
reliable tumor avidity on 18F-FDG-PET. Other right portal vein (Bismuth, 1982; Majno et al., 2014).
tracers such as gallium-68 tetraazacyclododecane Deep within the liver, hepatic arterial branches,
tetraacetic acid–octreotate (DOTATATE) are cur- portal–venous branches and biliary ducts run
rently in development to improve the sensitivity of alongside one another, enclosed in the Glissonian
PET or scintigraphy in these tumor types. sheath. The hepatic arterial anatomy is neverthe-
The liver tumor burden can be determined on less more complex and variable than the portal–
all three imaging modalities, and programs are venous anatomy, because anatomical variants of
being developed to segment the liver automati- the hepatic artery can occur on three different
cally based on thresholds for contrast enhance- levels: the origin of hepatic arterial branches, the
ment or metabolic activity. Some tumors replace branching pattern of the hepatic arterial tree, and
liver parenchyma and some add to the total liver the segmental territory vascularized by the indi-
volume. Guidelines warn against treating patients vidual hepatic arterial branches (van den Hoven
with liver tumor burden >50% or >70% to ensure et al., 2015). Anatomical variants of the portal vein,
enough healthy liver tissue reserve to tolerate on the contrary, are mainly limited to variants in
radioembolization; a measurement of volume and branching order (van Leeuwen et al., 1994).
function of liver tissue would be more valid, but is The standard arterial anatomy of the adult liver
not currently standard. is described as a common hepatic artery (CHA)
originating from a celiac trifurcation that gives
1.3.3 ASSESSING THE INDIVIDUAL off the gastroduodenal artery branch (GDA) and
HEPATIC ARTERIAL ANATOMY then continues as the proper hepatic artery (PHA),
dividing into the left (LHA) and right (RHA)
The functional anatomy of the liver is based on the hepatic arteries, vascularizing segments 2–4 and
branching pattern of the portal vein, which usu- 5–8, respectively. However, approximately half of
ally parallels the hepatic artery and biliary ducts. radioembolization candidates have a variant to
According to the Couinaud model of segmental this configuration (van den Hoven et al., 2015).
anatomy, eight (or nine in the Couinaud–Bismuth An explanation of why variants of the hepatic
model) liver segments can be distinguished with a arterial anatomy are so common lies in the embry-
distinct vascularization and biliary drainage. An ological development. During early development,
avascular plane, called the portal scissura, separates three main arteries exist: an embryological LHA
the functional left and right liver. Cantlie’s line—an (eLHA) originating from the left gastric artery
imaginary line drawn anteriorly from the middle (LGA), an embryological middle hepatic artery
of the gallbladder fossa to the inferior vena cava (eMHA) from the celiac axis, and an embryologi-
posteriorly—or the course of the middle hepatic cal right hepatic artery (eRHA) from the superior
1.3 Pretreatment workup / 1.3.4 Preparatory angiography and intraprocedural imaging 13

mesenteric artery (SMA). The eLHA and eRHA and following them up to their segmental territory
should eventually regress, so that the eMHA forms of vascularization, with attention for the order of
a separate LHA and RHA that take over the vas- branching. The fissure for the ligamentum venosum
cularization of the entire liver (Wang et al., 2010). and the portocaval space should be screened with
However, one (or both) of the embryological arter- special attention, since these are the locations where
ies often persists, resulting in the presence of a the majority of aberrant LHAs and aberrant or early
hepatic artery with an aberrant origin, a so-called branching RHAs, respectively, course before enter-
aberrant hepatic artery. Logically, most aberrant ing the liver (van den Hoven et al., 2014b, 2015).
hepatic arteries originate from the LGA or SMA. Chapter 3 discusses hepatic arterial anatomy in the
Aberrant hepatic arteries can be divided into context of treatment planning in additional detail.
replaced hepatic arteries that are the main supply
to the right or left hepatic lobe (or both in the case 1.3.4 PREPARATORY
of a replaced CHA), and accessory hepatic arter- ANGIOGRAPHY AND
ies that only supply a part of the lobe. In the latter INTRAPROCEDURAL IMAGING
case, the remaining part of the liver lobe is vascu-
larized by a normally derived counterpart of the Preparatory angiography is performed before
aberrant LHA/RHA. treatment to map the arterial anatomy, to allow
A variant in the order of arterial branching is prophylactic or redistributive coil embolization of
present in up to 20% of patients. In these patients, arterial branches if necessary to determine opti-
an early branching LHA/RHA may be present as mal catheter positioning and to administer a simu-
(part of) the LHA or RHA that originates proximal lation scout tracer. Intra-arterial access is gained
to the GDA. Another variant is a trifurcation (or through transcutaneous puncture, using the
quadrifurcation) of the CHA. This means that the Seldinger technique, either through a transfemo-
PHA is absent, so that the CHA branches into a ral or transradial approach. After securing the
GDA, LHA, RHA (and MHA). access site, a preshaped catheter is used to enter the
Variants in the segmental vascularization pat- source of the hepatic arterial vasculature (usually
tern include variants in the origin of the arteries the celiac axis). A standard end-hole microcatheter
vascularizing segments 1 and 4, as well as unex- is advanced over an atraumatic microguidewire for
pected vascular territories of aberrant hepatic further selectively catheterization.
arteries. The arterial feeding branch(es) to segment Intraprocedural imaging with digital subtrac-
4 may originate from the LHA, RHA, both, or from tion angiography (DSA) and C-arm cone beam CT
a separate origin of the CHA/PHA. Segment 1 may (CBCT) plays a pivotal role during the prepara-
in addition be vascularized by a branch originating tory angiography. DSA provides two-dimensional
from the segment 4 artery. images of the vasculature at a high spatial resolu-
All of the previous outlined anatomical variants tion, providing a high-resolution projectional map
can coincide, leading to complex and unexpected to guide the catheterization. Furthermore, cin-
individual hepatic arterial configurations and seg- ematic DSA images can be acquired during high
mental vascularization patterns. Failing to identify rate contrast administration with a power injec-
aberrant hepatic arteries may result in incomplete tor, thereby depicting arterial flow rates, tumor
treatment, whereas incorrect judgment of the seg- blushes, and altered dynamics.
mental vascularization pattern may result in under- CBCT is a relatively new imaging modality
or overdosing of the treatment activity (van den that enables the acquisition of three-dimensional
Hoven et al., 2014b). Thus, it is essential to assess the CT-like images, depicting contrast-enhanced ves-
individual hepatic arterial anatomy in each patient. sels in relation to their surrounding soft-tissue
Assessment on pretreatment arterial enhanced structures, by rotation of the C-arm mounted
CT or MRI enables identification and characteriza- flat-panel detector around the patient. The timing
tion of anatomical variants, while providing guid- between contrast injection and start of the scan
ance for subsequent catheterization during the can be adjusted to influence whether the images
preparatory angiography. Reading the pretreatment mainly depict contrast enhancement of the arte-
CT or MRI should be done systematically, scanning rial tree (early arterial phase), the liver and tumor
all potential sources for hepatic arterial branches, parenchyma (late arterial phase, Figure 1.2a and b),
14 Introduction to hepatic radioembolization

or both (van den Hoven et al., 2015b). The hepatic arteries (SDAs) deserve special attention. Nontarget
arterial anatomy is mapped by a combination of deposition into the cystic artery and the falciform
DSA and CBCT. “Complete” hepatic arteriography artery is less likely to be life threatening. The cath-
should be performed to depict comprehensively eter can often be positioned distal to these side
all arteries supplying any portion of the liver (Liu branches without sacrificing tumor coverage, but
et al., 2005; Lewandowski et al., 2007; Salem et al., this may require splitting of doses. If that is not pos-
2007; van den Hoven et al., 2014b). sible, prophylactic embolization with coils or vascu-
“Complete” hepatic angiography requires identi- lar plugs may be performed. Routine embolization
fication of parasitized extrahepatic arteries (Figure of the GDA was previously a standard of care but
1.3). These are tumor-feeding branches that have has become controversial, since this may actually
been recruited from arteries outside the hepatic vas- induce the hypertrophy of new, very small hepatof-
culature (including prominent phrenic, intercostal, ugal vessels during the interval between the prepa-
omental, internal mammary, adrenal, renal capsu- ratory angiography and treatment (Abdelmaksoud
lar, gastric, and pancreaticoduodenal arteries) by et al., 2010; Lam et al., 2013a; Samuelson et al.,
stimulation of neovascularization and are found in 2013). Alternatively, administration of yttrium
17% of radioembolization patients. These branches microspheres using an antireflux catheter or
can often be embolized, causing intrahepatic arter- occlusion balloon catheter may provide a solution
ies to take over the entire blood supply to the tumor, (Ahmadzadehfar et al., 2011; Prince et al., 2014b).
to ensure complete tumor coverage during treat- Choosing the optimal catheter position(s)
ment (Abdelmaksoud et al., 2011a). Failure to recog- requires attention for the risk of extrahepatic
nize these arteries results in incomplete treatment. shunting by evaluating the distance between cath-
Since extrahepatic deposition of radioactive eter tip and patent gastrointestinal side branches,
microspheres may cause serious complications, by evaluating tumor coverage by making sure
it should be assessed whether side branches of the that the catheter is placed proximal to all tumor-
hepatic arterial vasculature pose a risk. The GDA, feeding branches, and by evaluating flow dynam-
right gastric artery (RGA), and supraduodenal ics by recognizing preferential flow directions. In

(a) (b)

Figure 1.2 C-arm CT images demonstrating the arterial perfusion territory of the right (a) and left
(b) hepatic artery. These late arterial phase images were separately acquired, with contrast injection
by a microcatheter placed in the right and left hepatic artery, respectively. Both the healthy liver
parenchyma and the tumor in the posterior part of the right liver lobe show contrast enhancement.
Note how the vascular territories of both arteries complement each other.
1.3 Pretreatment workup / 1.3.5 Imaging of the scout dose distribution 15

(a) (b)

Figure 1.3 Parasitized left inferior phrenic artery. The arterial phase of the pretreatment CT (a) shows
a prominent left inferior phrenic artery coursing adjacent to the upper part of a large hypervascular
tumor mass (arrow). The suspected presence of a parasitized extrahepatic artery was confirmed dur-
ing angiography. Digital subtraction angiography acquired after selective catheterization of the left
inferior phrenic artery (b) demonstrates intrahepatic tumor blush (arrow).

general, it is considered impractical to use more 1.3.5 IMAGING OF THE SCOUT


than two to three injection positions. Coil embo- DOSE DISTRIBUTION
lization of intrahepatic arterial branches (e.g., the
S4A, or an accessory LHA) may be used as a means Currently, imaging of the scout dose distribution is
to reduce the number of required injection posi- mainly performed for safety reasons, to quantitate
tions, since this will induce redistribution of blood hepatopulmonary shunting and to rule out unin-
flow through intrahepatic collateral vessels (Bilbao tentional extrahepatic deposition. Planar scintigra-
et al., 2010; Abdelmaksoud et al., 2011b). phy can be used to calculate the lung shunt fraction
At the end of the preparatory angiography, it is after administration of 99mTc-MAA. For resin micro-
advisable to do a test injection with contrast agent spheres, the manufacturer recommends to lower the
from the intended treatment positions. A gentle treatment activity in patients with a lung shunt frac-
hand injection simulating the administration tion of 10%–20%, and to refrain from treatment in
injection may reveal problematic flow dynamics patients with a lung shunt fraction of >20%. This is
resulting in a disproportional microsphere distri- a convenient and simple recommendation based on
bution over tumor-bearing liver segments, while a whole liver treatment of average sized persons, but
late arterial phase CBCT may show unintentional with some critical flaws. First, lung shunt fraction
gastrointestinal shunting or lack of target segment is less important than the estimated lung-absorbed
perfusion (van den Hoven et al., 2015b). dose. The latter should be kept below 30 Gy (or 50
After confirming adequacy of the catheter Gy cumulatively in patients who undergo sequen-
position(s), a simulation tracer test dose of scout par- tial treatments). Second, planar scintigraphy-based
ticles is injected. Circa 30–150 MBq technetium-99m assessment gives a less accurate estimation of the
(99mTc)-macroaggregated albumin (MAA) is used as lung-absorbed dose than SPECT/CT, oftentimes
a surrogate for 90Y-microspheres, while a scout dose averaging hepatic dome activity into right lung base
(250 MBq) of identical 166Ho-microspheres is avail- activity (Yu et al., 2013). Third, it has been demon-
able for 166Ho-microspheres treatment. DSA images strated that the use of 99mTc-MAA itself leads to an
of the exact catheter position during injection should overestimation of the true liver-to-lung shunting,
be stored. probably due to differences in particle character-
The access site is closed by manual compression istics, the broad range of 99mTc-MAA particle size,
until hemostasis, or by using a closure device. and circulating free pertechnetate (Elschot et al.,
16 Introduction to hepatic radioembolization

2014; Smits et al., 2014). These points are discussed in method. According to this method, a fixed treat-
greater detail in Chapter 4. ment activity of 90Y between 2 and 3 GBq (depend-
SPECT/CT is considered the gold standard for ing on the tumor burden) is prescribed. This
detection of unintentional extrahepatic activity in method is no longer advocated, because it has
Europe. However, its relatively low spatial resolu- been associated with unacceptable clinical toxicity
tion and potential occurrence of misregistration (Smits et al., 2014).
between the SPECT data set and the low-dose CT The body surface area (BSA) method adjusts
data set can make the distinction between intra- the prescribed activity for the patient’s BSA and
and extrahepatic activity accumulation challenging. the fractional tumor burden. It assumes a corre-
Therefore, many centers now base their assessment lation between BSA and liver weight, but this is
of extrahepatic perfusion primarily on CBCT. not necessarily true for patients with liver cancer.
The intrahepatic scout dose distribution should As a consequence, small patients with a relatively
ideally be a good predictor for the treatment distri- large liver will be undertreated, potentially result-
bution, since determining the particle distribution ing in progressive disease, while tall patients with
in tumorous and nontumorous tissue (T/N ratio) a small cirrhotic liver will receive too much activ-
would enable a patient-tailored treatment strategy. ity, which may result in hepatotoxicity (Lam et al.,
However, it has been demonstrated that the intra- 2014). In the vast majority of cases, however, it is a
hepatic distribution of 99mTc-MAA is not a reliable safe method to use, but it may be overly conserva-
predictor for the 90Y-microsphere distribution, tive for tumor-enlarged livers. The manufacturer
especially not in patients with relatively hypovas- of resin 90Y-microspheres recommends the use of
cular liver tumors (Wondergem et al., 2013). This this method.
may be explained by the fact that the difference in The method based on medical internal radiation
particle characteristics, including different particle dose (MIRD) is recommended by the manufac-
size, density, shape, and number, strongly affects turer of glass 90Y-microspheres. With this method,
the particle distribution (Van de Wiele et al., 2012). the prescribed activity is determined by calcu-
In patients with markedly hypervascular HCCs, lating the activity required to achieve a desired
preferential tumoral blood flow is so strong that absorbed dose (usually 80–120 Gy), assuming a
these differences in particle characteristics have homogeneous intrahepatic microsphere distribu-
less influence, making the 99mTc-MAA distribution tion throughout the treated portion of the liver and
a more reliable predictor (Garin et al., 2012; Garin, a known yield of 50 Gy per GBq 90Y per kilogram
2015). of liver tissue. Although this method accounts for
Using a scout dose of particles identical liver mass, it neglects the inter- and intrapatient
to the treatment microspheres, such as with variability of intrahepatic microsphere distribu-
166
Ho-microspheres, may provide a more accu- tion between tumorous and nontumorous tis-
rate prediction of the intrahepatic therapeutic sue (T/N ratio). It does not account for fractional
microsphere distribution. Further investigation is or total tumor burden. The activity calculation
required to confirm this hypothesis, because dif- method for 166Ho-microspheres is also a MIRD
ferences in catheter positioning between the scout method, with a fixed target whole liver-absorbed
procedure and the therapy procedure may also play dose of 60 Gy (maximum tolerable dose derived
an important role. The embolic effect of a scout dose from a phase I trial) and the 166Ho-specific energy
could theoretically change flow patterns, and even yield of 15.87 Gy per GBq per kilogram.
the beginning and end of a partially embolic thera- An anatomic partition model could be the most
peutic administration could have different dynam- scientifically sound method for pretreatment activ-
ics (Prince et al., 2015; van den Hoven et al., 2015a). ity calculations. This method extends the MIRD
calculation by incorporating the T/N microsphere
1.3.6 PRETREATMENT ACTIVITY uptake ratio, the weight of the healthy liver tissue,
CALCULATIONS and the maximum tolerable healthy liver-absorbed
dose. Partition models are currently applicable for
Different methods for pretreatment activity cal- patients with few and well-defined tumors where a
culations (pretreatment dosimetry) have been T/N ratio can be estimated. However, partitioning
proposed. The empirical method is the simplest the liver into only two discreet volumes is at best
1.4 Treatment / 1.4.2 Treatment technique 17

inaccurate and in many cases impossible when biliary drainage, or biliary stent are at high risk for
disease is diffuse, infiltrative, and/or hypovascular. abscess formation and should be administered a
Physiologic and functional imaging methods are long course of prophylactic antibiotics before and
being developed to address this patient population. after the procedure.
Radioembolization treatment planning and
dosimetry techniques are discussed in greater 1.4.2 TREATMENT TECHNIQUE
detail in Chapter 5.
During treatment, radioactive microspheres
should be administered at the same catheter posi-
tion as during the preparatory angiography unless
1.4 trEatMENt contraindications were discovered from the scout
dose simulation. In patients with bilobar disease,
Most centers perform the preparatory angiography treatment of both lobes can either be performed at
and treatment sessions as outpatient procedures, once or in two-staged treatment sessions. The lat-
but some patients may require admission to the ter may be advisable in patients with a high tumor
hospital before and/or after undergoing treatment, burden to allow a more careful approach with
since clinical observation and drug therapy may regard to toxicity.
be required. The treatment procedure is typically It is advisable to perform a last check with DSA
scheduled ~1–2 weeks after a successful prepara- and/or CBCT before the administration, to make
tory angiography, but in some circumstances, can sure that no new hepatico-enteric collaterals have
even be performed on the same day as the prepara- been recruited in the interval between the prepara-
tory angiography. tory angiography and treatment, and that the cath-
eter is indeed placed in the same position.
1.4.1 MEDICATION AND Although the administration systems of the dif-
PERIPROCEDURAL CARE ferent microspheres slightly differ from each other,
they all have a vial that contains the radioactive
Monitoring of vital signs, with a heart rate moni- microspheres in an acrylic container, an affer-
tor, electrocardiogram (ECG) registration, pulse ent line that allows the operator to inject solution
oximetry, and periodic blood pressure measure- into the vial, and an efferent line that connects
ments, is considered standard. The use of proton the vial with the microcatheter. After setting up
pump inhibitors or H2 receptor antagonists dur- the administration system, ensuring that no air is
ing the first week before treatment and a month present in any of the lines, administration of the
after treatment is recommended as a prophylaxis microspheres can be started.
for gastrointestinal ulcer formation. Furthermore, A 5% glucose solution has replaced the formerly
corticosteroids given for a few days or weeks may recommended sterile water for injection of resin
help to mitigate the expected symptoms of postem- microspheres. Sterile water presumably leads to
bolization syndrome. Intravenous analgesia and temporary changes in blood osmolality, causing
sedation should also be considered during the hemolysis, vascular endothelial damage, vaso-
procedure to control pain and anxiety (Mahnken spasm, and premature stasis of blood flow during
et al., 2013). Heparin (up to 50 IU/kg) may be injection. Recently, it has been demonstrated that
administered intra-arterially or intravenously at using isotonic 5% glucose water instead reduces
the beginning of the procedure for thrombosis the occurrence of stasis, reduces periprocedural
prophylaxis. Intra-arterial administration of a pain, and improves the percentage delivered activ-
vasodilator (nitroglycerine or a calcium antago- ity (Ahmadzadehfar et al., 2015). Saline and iodin-
nist) is indicated if vasospasm occurs at any time ated contrast should not be used to avoid the risk of
during the procedure. Patients treated for NET displacement of the yttrium-90 from resin micro-
may require aggressive somatostatin analog pro- spheres via an ion exchange mechanism.
phylaxis against carcinoid crisis from sudden hor- The injection technique differs signifi-
monal release. Patients with enterobiliary reflux cantly between glass 90Y-microspheres and resin
90
from prior surgery, endoscopic retrograde chol- Y-microspheres or 166Ho-microspheres due
angiopancreatography (ERCP), sphincterotomy, to the significantly lower number of injected
18 Introduction to hepatic radioembolization

microspheres. Glass microspheres can be injected with the vessel wall during administrations to
in a single bolus, without angiographic monitoring, prevent particle reflux without blocking antegrade
since stasis of blood flow or reflux is not expected. blood flow. Using this ARC makes embolization
A typical injection with glass microspheres may of side branches proximal to the administration
be completed in less than 5 min. When using site unnecessary (Fischman et al., 2014; Morshedi
resin 90Y-microspheres or 166Ho-microspheres, et al., 2014; van den Hoven et al., 2014a). Earlier
injection should be, however, performed care- studies also used occlusion balloons for adminis-
fully, using short pulsatile pressure strokes on tration, with similar objectives.
the syringe (ideally a small syringe of 3–5 cc). The Using the ARC may in theory also affect par-
blood flow velocity should be regularly checked, ticle distribution in two ways. First, the fixed posi-
since the administration should be paused in slow tion of the ARC may improve the predictive value
flow conditions, and stopped entirely when sta- of the treatment simulations with a scout dose par-
sis occurs to prevent particle reflux. A change in ticle (99mTc-MAA or 166Ho) by preventing catheter
preferential blood flow direction can sometimes be tip deviation. Second, the design of the ARC has a
observed during the administration, which may complex effect on blood flow and particle outflow
be explained by stasis in some branches and sub- dynamics, which may ultimately improve tumor
sequent redistribution of blood flow toward other targeting (Pasciak et al., 2015; van den Hoven et al.,
branches. The treatment is completed if the entire 2015a).
activity is administered, or when the administra-
tion is stopped prematurely for stasis, which can 1.4.4 IMAGING OF THE
take up to 30 min in total. THERAPEUTIC MICROSPHERE
To determine the net administered activity, the DISTRIBUTION
vial, administration lines, and catheter may be
checked for remnant activity with a dose calibra- Imaging of the therapeutic microsphere distribu-
tor. The process of pre- and posttreatment assay of tion is increasingly performed. Bremsstrahlung
administered activity and residual is described in SPECT/CT was formerly the only technique avail-
Chapter 7. able to depict the 90Y distribution after treatment,
and was limited by poor spatial resolution. More
1.4.3 CATHETER TYPES AND recently, internal-pair production-based PET/CT
PARTICLE-FLUID DYNAMICS has largely replaced bremsstrahlung SPECT/CT
due to its superior spatial resolution and lower
Two types of administration catheters are commer- scatter (Padia et al., 2013; Elschot et al., 2013b). The
cially available for radioembolization procedures. biodistribution of 166Ho-microspheres can also be
The standard end-hole microcatheter remains the imaged by SPECT, using the primary γ-radiation
standard default catheter for all embolotherapies emission, as well as by MRI, by measuring the R2∗
in interventional radiology. This may be explained dephasing effects induced by the paramagnetic
by the low cost, simplicity of use, atraumatic char- microspheres (Elschot et al., 2013a; Smits et al.,
acter, and wide range of available sizes and flexibil- 2013a; van de Maat et al., 2013).
ity that allow for catheterization of small, tortuous Confirming an adequate intrahepatic treat-
vessels. However, a disadvantage is that the cath- ment distribution as well as lack of extrahepatic
eter has no fixed support in the vessel lumen. activity distribution seems a logical part of treat-
Therefore, the microcatheter may deviate toward a ing patients with radioembolization. However,
vessel wall during injection of microspheres, which this has not yet become standard of care. One of
can lead to streaming and preferential deposition the reasons is lack of financial reimbursement.
in daughter branches on the side of deviation. Furthermore, low signal-to-noise ratio on these
A microcatheter specifically designed for embo- images may limit the accuracy of both visual and
lotherapy, the Surefire Infusion System (Surefire quantitative assessments of the 90Y-activity on
Medical Inc., Westminster, Colorado), has been PET/CT (Pasciak et al., 2014). Also, the clinical
developed to prevent reflux during radioembo- consequences of finding an unfavorable micro-
lization. The tip of this antireflux catheter (ARC) sphere distribution on posttreatment imaging
dynamically expands radially to make contact remain uncertain and irreversible. Nevertheless,
1.4 Treatment / 1.4.5 Dose–response relationship 19

90
Y-PET/CT and 166Ho-SPECT/CT should be that there is high intraindividual dose-distribution
incorporated into study protocols of future clini- heterogeneity and that inadequate treatment of at
cal trials on radioembolization to gather data least one tumor is fairly common in patients with
about dosimetry and treatment outcomes. While colorectal cancer liver metastases (Flamen et al.,
such data could beneficially impact radioemboli- 2008; Smits et al., 2013a). Dose–response relation-
zation in the future, the methods by which these ships for radioembolization will be discussed in
data are gathered must be carefully controlled as greater detail in Chapter 8.
described in Chapter 15. If effective tumor-absorbed dose thresholds can
be established for the various microspheres and
tumors, and imaging can monitor actual distribu-
tion of therapeutic dose, an iterative administration
1.4.5 DOSE–RESPONSE technique should be feasible. Additional treatment
RELATIONSHIP could be given to tumors that appear to have received
inadequate initial treatment. If feasible, such a strat-
Several studies have shown an association between egy should translate into improved outcomes.
tumor-absorbed dose and tumor response and
overall survival after radioembolization. Therefore,
the goal is to strive for a high tumor-absorbed dose
(Figure 1.4). However, a clear threshold or goal for 1.5 trEatMENt-rELatED
effective tumor-absorbed dose has not been found. LaBOratOrY aND CLINICaL
Most dose–response investigations have been tOXICItY
performed in patients treated with HCC with
glass 90Y-microspheres (Strigari et al., 2010; Riaz et 1.5.1 COMPLAINTS DURING
al., 2011; Garin et al., 2012, 2015; Kao et al., 2013;
TREATMENT
Mazzaferro et al., 2013; Eaton et al., 2014; Srinivas
et al., 2014). These dose–response relationships Microsphere administration may sometimes
may not apply to other cell types and microsphere induce complaints such as abdominal discomfort,
types (Flamen et al., 2008; Lam et al., 2013b, 2015; pain, nausea, and vomiting. This can be accom-
Demirelli et al., 2015). The difference in tumor biol- panied by a vasovagal reaction. The exact cause
ogy among various tumors may require a different of these symptoms remains unclear, but clinical
amount of radiation to induce tumor cell death, observation suggests that complaints often occur
similar to findings in EBRT (Lausch et al., 2013). simultaneously with stasis of blood flow, indicat-
Furthermore, due to activity and distribution dif- ing that the embolic effect of therapy is causing
ferences between resin and glass 90Y-microspheres, symptoms. The symptoms are self-limited and can
generally higher tumor-absorbed doses are last for days but usually resolve on the day of treat-
reported for glass microspheres, without a signifi- ment with supportive medical therapy.
cantly different biological effect (Cremonesi et al.,
2014). 1.5.2 LABORATORY TOXICITY
Some authors report that an absorbed dose of
120 Gy is commonly considered tumoricidal for Abnormal laboratory values (liver function tests,
HCC (Riaz et al., 2011), whereas colorectal can- complete blood count) should be expected for sev-
cer liver metastases may require at least 70 Gy eral weeks after treatment. Mild-to-severe (CTCAE
(Srinivas et al., 2014). Yet, a recent literature review grades 3–4) laboratory toxicity may even occur in
showed that the range of reported effective tumor- one-third of patients without signs of associated
absorbed dose values is extremely wide, with clinical toxicity (Smits et al., 2013b). Therefore, labo-
66–495 Gy for resin microspheres and 163–1214 ratory investigations are not suited to discriminate
Gy for glass microspheres. The differences in study between patients with and without a normal reaction
population and methods for quantifying tumor to treatment. Abnormal liver function test (espe-
dose and response in these studies certainly con- cially bilirubin level) in combination with ascites,
tribute to this uncertainty. Even within an individ- however, is an alarming finding that may indicate
ual liver, two studies independently demonstrated radioembolization-induced liver disease (REILD).
(a) (b) (c) (d) (e)
20 Introduction to hepatic radioembolization

(f ) (g) (h) (i) (j)

Figure 1.4 Clinical examples of colorectal carcinoma liver metastasis patients with adequate and inadequate tumor targeting. The upper row (a–e)
shows a patient with a large tumor mass on pretreatment T2-weighted (T2W) MRI (a) and 18F-FDG-PET (b) that extends from the central liver to the
left liver lobe, which did not show a sufficient tumor-absorbed dose on 90Y-PET/CT (c). At 1-month posttreatment, progressive disease was noted on
the MRI (d) and 18F-FDG-PET (h). The patient in the lower row (f–j), on the contrary, had a metastasis in the posterior sector of the right liver lobe
(f and g) that did have a sufficient tumor-absorbed dose on 90Y-PET/CT (h). The posttreatment MRI (i) at 1-month follow-up showed reduction in size
and 18F-FDG-PET (j) demonstrated a complete metabolic response.
1.6 Tumor response assessment / 1.6.1 Anatomical tumor response assessment 21

1.5.2.1 Postembolization syndrome may lead to the development of REILD within


the first 2 months after treatment. Clinically, it
The most common symptoms after radioemboli- is characterized by jaundice, weight gain, ascites,
zation are fatigue, abdominal pain, nausea, vom- and a distinct rise in bilirubin, while transami-
iting, low-grade fever, and anorexia/cachexia. nases and alkaline phosphatase are only mildly
These symptoms constitute the so-called post- increased. Pathologically, REILD is characterized
radioembolization syndrome, which is related by sinusoidal congestion, venule occlusion by
to complaints after other embolotherapies (Riaz sloughed necrotic endothelium, and eventually
et al., 2009). These symptoms are considered an fibrosis of the liver. If severe, REILD can be fatal
expected reaction to radioembolization treat- in the acute phase, but some patients with milder
ment and they are usually self-limited and REILD may develop chronic hepatic insufficiency
manageable with standard supportive medical and complications of portal hypertension. The
treatment. Failure to recognize this may unfortu- radiation tolerance of the liver tissue depends
nately result in unnecessary extension of patient on the involved volume, previous exposure to
hospitalization. hepatotoxins including many common systemic
chemotherapeutics, and underlying cirrhosis
(Gil-Alzugaray et al., 2013). The diagnosis and
1.5.2.2 Treatment complications management of complications following radio-
embolization are discussed in greater detail in
Treatment complications are uncommon, but Chapter 14.
can occur by accumulation of radioactive micro-
spheres outside the liver or overexposure of the
healthy liver tissue to radiation.
Extrahepatic activity in the gallbladder wall,
pancreas, and gastrointestinal tract may, respec-
1.6 tUMOr rESPONSE
tively, result in radiation-induced cholecysti- aSSESSMENt
tis, pancreatitis, or gastric/duodenal ulceration.
Radiation-induced cholecystitis and pancreatitis A variety of methods can be used to assess tumor
are often subclinical in presentation, but may in response after radioembolization treatment.
some cases cause severe morbidity. Radiation- These methods can be divided into morpho-
induced ulcers are particularly cumbersome logic and physiologic tumor response assessment
because they originate from the serosal surface methods. Morphologic or anatomical assess-
instead of the mucosal surfaces as in normal ment methods only evaluate the treatment effect
peptic ulcers, impairing the healing process and on tumor size/volume as visualized on cross-
complicating surgical intervention. Detecting sectional imaging. Physiologic or functional
and correcting extrahepatic shunting before assessment methods assess the treatment effect
treatment can prevent these complications (Riaz on tumor biology, including arterial vasculariza-
et al., 2009). tion, water diffusivity, and glucose uptake (Figure
Accumulation of too much radioactive micro- 1.5). All of these methods strive to provide valu-
spheres in the lung causes tissue damage due to the able prognostic information using a reproducible
combined effects of ischemia and radiation. Cases method. In general, morphologic tumor assess-
of radiation pneumonitis have been described in ment is simple, standardized, relatively subjec-
the literature, many of them fatal, often associated tive, variably reproducible, and time consuming.
with hepatic venous and/or portal venous tumor Physiologic tumor assessments, on the other
invasion. To prevent this, estimated lung-absorbed hand, are complex, objective, and partly auto-
doses should be kept <30 Gy (or 50 Gy cumula- mated, but reproducibility is heavily dependent
tively) (Leung et al., 1995). Additional exposure to on the technique for image acquisition, recon-
pulmonary toxins such as chemotherapy may also struction, and analysis. Unfortunately, only few
compound the risk. comparative studies have been performed to
Exposing the healthy liver tissue to high radi- assess which methods work best in radioembo-
ation-absorbed doses during radioembolization lization patients.
22 Introduction to hepatic radioembolization

(a) (b) (c)

Figure 1.5 Early detection of tumor recurrence on functional imaging. This patient with colorec-
tal cancer liver metastasis had a good response after initial treatment with radioembolization, but
developed tumor recurrence in segment 4, 9 months after treatment. The metastasis is hardly visible
on a standard (T2W) MRI sequence (a), but was detected on diffusion-weighted (DW)-MRI (b) and on
18
F-FDG-PET (c). The patient received additional treatment of segment 4, based on this finding, and
responded well to treatment again.

1.6.1 ANATOMICAL TUMOR There are no guidelines on the required time


RESPONSE ASSESSMENT intervals for follow-up imaging, but most oncol-
ogy studies adapt follow-up to chemotherapy
WHO published the first response criteria in 1981. treatment cycles (e.g., in intervals of 3 weeks) and
These criteria use the sum of the product (SPD) interventional radiology treatments often use 1,
of tumor diameters (longest diameter in the axial 2, and/or 3 months posttreatment. If scans are
imaging plane and the one perpendicular to it) to performed with short intervals (i.e., <6 weeks),
evaluate whether therapy led to significant tumor parameters such as time to progression (TTP)
shrinkage. Response was classified into complete or progression-free survival (PFS) may be used
response (CR, –100%), partial response (PR, –50% (Llovet et al., 2008).
to –99%), stable disease (SD, –49% to 25%), and RECIST has been adopted as the standard for
progressive disease (PD, >25%), based on the per- response assessment by both research institu-
centage of change in SPD. tions and regulatory authorities. It functions as a
The Response Evaluation Criteria in Solid Tumors surrogate endpoint for overall survival, provides
(RECIST) system was introduced to simplify the important prognostic information (especially
evaluation, and was found to correlate highly with in therapies where tumor shrinkage is expected
the WHO system, and version 1.1 is now the most such as cytotoxic chemotherapy), is relatively
widely used response classification system in solid simple and reproducible, and does not require
tumors. It uses the sum of the longest one-dimen- state-of-the-art imaging facilities. However,
sional tumor diameters in two target lesions on its validity is questioned for therapies where
cross-sectional imaging to classify response into the response to therapy is not always associated with
same categories as the WHO criteria (CR –100%, lesion shrinkage, such as radioembolization,
PR –99% to –30%, SD –30% to 20%, PD >20%). ablation, and some systemic treatments such as
Target lesion response can also be extended to liver immunotherapy.
response and overall (whole-body) response by
incorporating information about the size change in 1.6.2 FUNCTIONAL TUMOR
nontarget lesions and the appearance of new lesions RESPONSE ASSESSMENT
in or outside the liver (Tirkes et al., 2013). Studies
generally report either the best response during Modern imaging technology has enabled a more
follow-up or the objective response rate (CR + PR) physiological approach to tumor response assess-
or disease control rate (CR + PR + SD) at a specific ment. Modified RECIST (mRECIST) has been
time of follow-up. developed to address the issue that a change in
References 23

arterial tumor vascularization might be more


representative than a change in entire tumor size 1.7 CONCLUSION
in patients with HCC treated with locoregional
therapy. Similar to RECIST, the longest diam- In recent decades, radioembolization has evolved
eter is measured but only of the viable (arterially into a safe and effective liver cancer therapy.
enhanced) portion of the tumor. Lack of arte- Despite major advances brought by modern imag-
rial tumor enhancement on follow-up imaging, ing technology and clinical experience, improve-
even if the total lesion size is unchanged or even ment of patient workup, treatment technique,
increased, is considered a complete response toxicity, and response assessment is an ongoing
(CR) (Lencioni and Llovet, 2010). The other process. In the following chapters, these subjects
response categories are defined similarly as with will be discussed in further detail.
RECIST based on the enhanced tumor diameter
measurement. REFERENCES
More quantitative approaches are offered by
dynamic contrast-enhanced MRI (DCE-MRI). Abdelmaksoud, M.H.K., Hwang G.L., Louie
With DCE-MRI, serial images are acquired before, J.D. et al. (2010). Development of new
during, and after contrast agent administra- hepaticoenteric collateral pathways
tion, allowing for contrast kinetic modeling with after hepatic arterial skeletonization in
time-signal intensity curves (Choyke et al., 2003). preparation for yttrium-90 radioemboliza-
MRI can also be used to assess functional tumor tion. J Vasc Interv Radiol 21:1385–1395.
response by diffusion-weighted imaging (DWI). Available at http://dx.doi.org/10.1016/j.
The principle behind this imaging technique is jvir.2010.04.030.
that the water diffusivity is restricted in tumors, as Abdelmaksoud, M.H.K., Louie, J.D., Kothary,
opposed to healthy liver tissue. Diffusivity can be N. et al. (2011a). Embolization of parasit-
quantified by calculating apparent diffusion coeffi- ized extrahepatic arteries to reestablish
cients (ADC). An increase of ADC after treatment intrahepatic arterial supply to tumors before
indicates treatment response (Barabasch et  al., yttrium-90 radioembolization. J Vasc Interv
2015). Radiol 22:1355–1362. Available at http://
18
F-FDG-PET is also increasingly used to www.ncbi.nlm.nih.gov/pubmed/21961979
assess treatment response. Standardized uptake [Accessed August 4, 2014].
values (SUV) can be used to quantify the selec- Abdelmaksoud, M.H.K., Louie, J.D., Kothary,
tive uptake of the 18F-isotope labeled glucose N. et al. (2011b). Consolidation of hepatic
analog in malignant tissues (Larson et al., 1999). arterial inflow by embolization of vari-
A combination of SUV and metabolic tumor ant hepatic arteries in preparation for
volume—called tumor lesion glycolysis or meta- yttrium-90 radioembolization. J Vasc Interv
bolic product—may be especially interesting, Radiol 22:1364–1371.e1. Available at http://
since this reflects the total glucose turnover in a www.ncbi.nlm.nih.gov/pubmed/21961981
tumor. Many options are available when acquir- [Accessed August 5, 2014].
ing, reconstructing, and analyzing PET data, yet Ahmadzadehfar, H., Meyer, C., Pieper, C.C. et al.
none of these methods has proven to be clearly (2015). Evaluation of the delivered activity of
superior to the others, and they may lead to dif- yttrium-90 resin microspheres using sterile
ferent results (Boellaard, 2011). An attempt to water and 5% glucose during administra-
standardize response analysis, PET Response tion. Eur J Nucl Med Mol Imaging Res 5:54.
Criteria in Solid Tumors (PERCIST) has not Available at http://www.ejnmmires.com/
resulted in the same widespread adaptation as content/5/1/54.
with the RECIST criteria. Ahmadzadehfar, H., Möhlenbruch, M., Sabet, A.
Both the anatomical and functional assess- et al. (2011). Is prophylactic embolization of
ments of tumor response following radioem- the hepatic falciform artery needed before
bolization are discussed in greater detail in radioembolization in patients with 99mTc-
Chapter 14. MAA accumulation in the anterior abdominal
24 Introduction to hepatic radioembolization

wall? Eur J Nucl Med Mol Imaging 38:1477– Braat, A.J.A.T., Smits, M.L.J., Braat, M.N.G.J.
1484. Available at http://www.ncbi.nlm.nih. et al. (2015). 90Y hepatic radioembolization:
gov/pubmed/21494857 [Accessed August 4, An update on current practice and recent
2014]. developments. J Nucl Med 56:1079–1087.
Ariel, I.M. (1965). Treatment of inoperable Available at http://jnm.snmjournals.org/cgi/
primary pancreatic and liver cancer by the doi/10.2967/jnumed.115.157446.
intra-arterial administration of radioactive Caldarola, L., Rosa, U., Badellino, F. et al. (1964).
isotopes (Y90 radiating microspheres. Ann Preparation of 32-P labelled resin micro-
Surg 162:267–278. spheres for radiation treatment of tumours
Ariel, I.M., Padula, G. (1982). Treatment of by intra-arterial injection. Minerva Nucl
asymptomatic metastatic cancer to the liver 55:169–174.
from primary colon and rectal cancer by the Campbell, A.M., Bailey, I.H., Burton, M.A. (2001).
intraarterial administration of chemotherapy Tumour dosimetry in human liver following
and radioactive isotopes. J Surg Oncol hepatic yttrium-90 microsphere therapy. Phys
20:151–156. Med Biol 46:487–498. Available at http://
Barabasch, A., Kraemer, N.A., Ciritsis, A. et al. www.ncbi.nlm.nih.gov/pubmed/11229728.
(2015). Diagnostic accuracy of diffusion- Choyke, P.L., Dwyer, A.J., Knopp, M.V. (2003).
weighted magnetic resonance imaging versus Functional tumor imaging with dynamic
positron emission tomography/computed contrast-enhanced magnetic resonance
tomography for early response assessment imaging. J Magn Reson Imaging 17:509–520.
of liver metastases to Y90-radioembolization. Available at http://www.ncbi.nlm.nih.gov/
Invest Radiol 50:409–415. pubmed/12720260. [Accessed August 4,
Bierman, H.R., Byron, R.L., Kelley, K.H., Grady, A. 2014].
(1951). Studies on the blood supply of tumors Cremonesi, M., Chiesa, C., Strigari, L. et al.
in man. III. Vascular patterns of the liver by (2014). Radioembolization of hepatic lesions
hepatic arteriography in vivo. J Natl Cancer from a radiobiology and dosimetric perspec-
Inst 12:107–131. tive. Front Oncol 4:210. Available at http://
Bismuth, H. (1982). Surgical anatomy and ana- journal.frontiersin.org/Journal/10.3389/
tomical surgery of the liver. World J Surg 6: fonc.2014.00210/. [Accessed September 10,
3–9. Available at http://www.ncbi.nlm.nih.gov/ 2014].
pubmed/7090393. D’Arienzo, M. (2013). Emission of β+ particles via
Blanchard, R.J., Grotenhuis, I., Lafave, J.W., internal pair production in the 0+ – 0+ transi-
Perry, J.F. (1965a). Blood supply to hepatic V2 tion of 90Zr: Historical background and cur-
carcinoma implants as measured by radioac- rent applications in nuclear medicine imaging.
tive microspheres. Proc Soc Exp Biol Med Atoms 1:2–12. Available at http://www.mdpi.
118:465–468. com/2218-2004/1/1/2/.
Blanchard, R.J., Lafave, J.W., Kim, Y.S. et al. Demirelli, S., Erkilic, M., Oner, A.O. et al.
(1965b). Treatment of patients with advanced (2015). Evaluation of factors affecting tumor
cancer utilizing Y90 microspheres. Cancer response and survival in patients with primary
18:375–380. and metastatic liver cancer treated with
Boellaard, R. (2011). Need for standardiza- microspheres. Eur J Gastroenterol Hepatol:
tion of 18F-FDG PET/CT for treatment 1. Available at http://content.wkhealth.com/
response assessments. J Nucl Med 52(Suppl linkback/openurl?sid=WKPTLP:landingpage
2):93S–100S. Available at http://www.ncbi. &an=00042737-900000000-98792.
nlm.nih.gov/pubmed/22144561 [Accessed Devcic, Z., Rosenberg, J., Braat, A.J. et al. (2014).
July 17, 2014]. The efficacy of hepatic 90Y resin radioem-
Braat, A.J.A.T., Huijbregts, J.E., Molenaar, I.Q. bolization for metastatic neuroendocrine
et al. (2014). Hepatic radioembolization as a tumors: A meta-analysis. J Nucl Med 55:1404–
bridge to liver surgery. Front Oncol 4:1–13. 1410. doi: 10.2967/jnumed.113.135855.
Available at http://journal.frontiersin.org/ Available at http://www.ncbi.nlm.nih.gov/
article/10.3389/fonc.2014.00199/abstract. pubmed/25012459 [Accessed July 18, 2014].
References 25

Eaton, B.R., Kim, H.S., Schreibmann, E. et al. microspheres. Phys Med Biol 53:6591–6603.
(2014). Quantitative dosimetry for yttrium-90 Available at http://www.ncbi.nlm.nih.gov/
radionuclide therapy: Tumor dose predicts pubmed/18978442 [Accessed August 5,
fluorodeoxyglucose positron emission 2014].
tomography response in hepatic metastatic Forner, A., Gilabert, M., Bruix, J., Raoul, J.-L.
melanoma. J Vasc Interv Radiol 25:288–295. (2014). Treatment of intermediate-stage
Available at http://dx.doi.org/10.1016/j. hepatocellular carcinoma. Nat Rev Clin
jvir.2013.08.021. Oncol 11:525–535. Available at http://dx.doi.
Elschot, M., Nijsen, J.F.W., Lam, M.G.E.H. org/10.1038/nrclinonc.2014.122 [Accessed
et al. (2014). (99m)Tc-MAA overestimates August 6, 2014].
the absorbed dose to the lungs in radio- Fuss, M., Salter, B.J., Herman, T.S., Thomas,
embolization: A quantitative evaluation in C.R. (2004). External beam radiation therapy
patients treated with 166Ho-microspheres. for hepatocellular carcinoma: Potential of
Eur J Nucl Med Mol Imaging 41:1965–1975. intensity-modulated and image-guided
Available at http://www.ncbi.nlm.nih.gov/ radiation therapy. Gastroenterology
pubmed/24819055 [Accessed January 8, 127:206–217.
2015]. Gaba, R.C., Lewandowski, R.J., Kulik, L.M. et al.
Elschot, M., Smits, M.L.J., Nijsen, J.F.W. et al. (2009). Radiation lobectomy: Preliminary
(2013a). Quantitative Monte Carlo-based findings of hepatic volumetric response to
holmium-166 SPECT reconstruction. Med Phys lobar yttrium-90 radioembolization. Ann
40:112502. Available at http://www.ncbi.nlm. Surg Oncol 16:1587–1596. Available at http://
nih.gov/pubmed/24320461 [Accessed August www.ncbi.nlm.nih.gov/pubmed/19357924
4, 2014]. [Accessed August 4, 2014].
Elschot, M., Vermolen, B.J., Lam, M.G.E.H. et al. Garin, E. (2015). Radioembolization with
(2013b). Quantitative comparison of PET and 90Y-loaded microspheres: High clinical impact
bremsstrahlung SPECT for imaging the in of treatment simulation with MAA-based
vivo yttrium-90 microsphere distribution after dosimetry. Eur J Nucl Med Mol Imaging
liver radioembolization. PLoS One 8:e55742. 42:1189–1191. Available at http://link.springer.
Available at http://www.pubmedcentral.nih. com/10.1007/s00259-015-3073-y.
gov/articlerender.fcgi?artid=3566032&tool= Garin, E., Lenoir, L., Rolland, Y. et al. (2012).
pmcentrez&rendertype=abstract [Accessed Dosimetry based on 99mTc-macroaggregated
July 24, 2014]. albumin SPECT/CT accurately predicts tumor
Emami, B., Lyman, J., Brown, A. et al. (1991). response and survival in hepatocellular carci-
Tolerance of normal tissue to therapeu- noma patients treated with 90Y-loaded glass
tic irradiation. Int J Radiat Oncol Biol Phys microspheres: Preliminary results. J Nucl Med
21:109–122. 53:255–263.
Eriksson, D., Stigbrand, T. (2010). Radiation- Garin, E., Rolland, Y., Edeline, J. et al. (2015).
induced cell death mechanisms. Tumor Biol. Personalized dosimetry and intensification
31: 363–372. concept with 90Y-loaded glass microsphere
Fischman, A.M., Ward, T.J., Patel, R.S. et al. radioembolization induce prolonged overall
(2014). Prospective, randomized study survival in hepatocelluar carcinoma patients
of coil embolization versus surefire infu- with portal vein thrombosis. J Nucl Med
sion system during yttrium-90 radioem- 56:339–346. Available at http://www.ncbi.nlm.
bolization with resin microspheres. J Vasc nih.gov/pubmed/25678490.
Interv Radiol. 25:1709–1716. doi: 10.1016/j. Giammarile, F., Bodei, L., Chiesa, C. et al. (2011).
jvir.2014.08.007. EANM procedure guideline for the treatment
Flamen, P., Vanderlinden, B., Delatte, P. et al. of liver cancer and liver metastases with intra-
(2008). Multimodality imaging can predict arterial radioactive compounds. Eur J Nucl
the metabolic response of unresectable Med Mol Imaging 38:1393–1406. Available
colorectal liver metastases to radioemboliza- at http://www.ncbi.nlm.nih.gov/pubmed/
tion therapy with yttrium-90 labeled resin [Accessed July 17, 2014].
26 Introduction to hepatic radioembolization

Gil-Alzugaray, B., Chopitea, A., Iñarrairaegui, M. Nucl Med Mol Imaging Res 3:57. Available at
et al. (2013). Prognostic factors and preven- http://www.pubmedcentral.nih.gov/articleren-
tion of radioembolization-induced liver dis- der.fcgi?artid=3733999&tool=pmcentrez&ren
ease. Hepatology 57:1078–1087. Available at dertype=abstract [Accessed August 4, 2014].
http://doi.wiley.com/10.1002/hep.26191. Kennedy, A. (2014). Radioembolization of hepatic
Grady, E.D. (1979). Internal radiation therapy of tumors. J Gastrointest Oncol 5:178–189.
hepatic cancer. Dis Colon Rectum 22:371–375. Available at http://www.pubmedcentral.nih.
Grady, E.D., Nolan, T.R., Crumbley, A.J. et al. gov/articlerender.fcgi?artid=4074949&tool=
(1975). Internal hepatic radiotherapy: II. pmcentrez&rendertype=abstract [Accessed
Intra-arterial radiocolloid therapy for hepatic July 23, 2014].
tumors. Am J Roentgenol Radium Ther Nucl Kennedy, A.S., Ball, D., Cohen, S.J. et al. (2015).
Med 124:596–599. Multicenter evaluation of the safety and
Grady, E.D., Sale, W., Nicolson, W.P., Rollins, L.C. efficacy of radioembolization in patients with
(1960). Intra-arterial radioisotopes to treat unresectable colorectal liver metastases
cancer. Am Surg 26:678–684. selected as candidates for 90 Y resin micro-
Grady, E.D., Sale, W.T., Rollins, L.C. (1963). spheres. J Gastrointest Oncol 90:134–142.
Localization of radioactivity by intravascular Kennedy, A.S., Nutting, C., Coldwell, D. et al.
injection of large radioactive particles. Ann (2004). Pathologic response and microdosim-
Surg 157:97–114. etry of (90)Y microspheres in man: Review of
Gray, B., Van Hazel, G., Hope, M. et al. (2001). four explanted whole livers. Int J Radiat Oncol
Randomised trial of SIR-Spheres plus chemo- Biol Phys 60:1552–1563. Available at http://
therapy vs. chemotherapy alone for treating www.ncbi.nlm.nih.gov/pubmed/15590187
patients with liver metastases from primary [Accessed August 1, 2014].
large bowel cancer. Ann Oncol 12:1711–1720. Kim, Y.S., Lafave, J.W., Maclean, L.D. (1962). The
Available at http://www.ncbi.nlm.nih.gov/ use of radiating microspheres in the treat-
pubmed/11843249 [Accessed August 2, 2014]. ment of experimental and human malignancy.
Gray, B.N., Burton, M.A., Kelleher, D.K. et al. Surgery 52:220–231.
(1989). Selective internal radiation (SIR) Lam, M.G.E.H., Banerjee, A., Goris, M.L. et al.
therapy for treatment of liver metastases: (2015). Fusion dual-tracer SPECT-based
Measurement of response rate. J Surg Oncol hepatic dosimetry predicts outcome after
42:192–196. radioembolization for a wide range of tumour
Gulec, S.A., Siegel, J.A. (2007). Posttherapy cell types. Eur J Nucl Med Mol Imaging
radiation safety considerations in radiomicro- 42:1192–1201. Available at http://www.ncbi.
sphere treatment with 90Y-microspheres. J nlm.nih.gov/pubmed/25916740.
Nucl Med 48:2080–2086. Lam, M.G.E.H., Banerjee, S., Louie, J.D. et al.
Hilgard, P., Hamami, M., Fouly, A.E.L. et al. (2010). (2013a). Root cause analysis of gastroduo-
Radioembolization with yttrium-90 glass denal ulceration after yttrium-90 radioem-
microspheres in hepatocellular carcinoma: bolization. Cardiovasc Intervent Radiol
European experience on safety and long-term 36:1536–1547. Available at http://www.ncbi.
survival. Hepatology 52:1741–1749. nlm.nih.gov/pubmed/23435742 [Accessed
Jernigan, S.R., Osborne, J.A., Mirek, C.J., August 4, 2014].
Buckner, G. (2015). Selective internal radiation Lam, M.G.E.H., Goris, M.L., Iagaru, A.H. et al.
therapy: Quantifying distal penetration and (2013b). Prognostic utility of 90Y radioem-
distribution of resin and glass microspheres bolization dosimetry based on fusion 99mTc-
in a surrogate arterial model. J Vasc Interv macroaggregated albumin-99mTc-sulfur
Radiol 26:897–904. Available at http://www. colloid SPECT. J Nucl Med 54:2055–2061.
ncbi.nlm.nih.gov/pubmed/25891507. Available at http://www.ncbi.nlm.nih.gov/
Kao, Y.-H., Steinberg, J.D., Tay, Y.S. et al. (2013). pubmed/24144563 [Accessed August 4, 2014].
Post-radioembolization yttrium-90 PET/CT Lam, M.G.E.H., Louie, J.D., Abdelmaksoud,
- part 2: Dose-response and tumor predic- M.H.K. et al. (2014). Limitations of body
tive dosimetry for resin microspheres. Eur J surface area-based activity calculation for
References 27

radioembolization of hepatic metastases Cancer Inst 100:698–711. Available at http://


in colorectal cancer. J Vasc Interv Radiol www.ncbi.nlm.nih.gov/pubmed/18477802
25:1085–1093. Available at http://www.ncbi. [Accessed July 11, 2014].
nlm.nih.gov/pubmed/24457263 [Accessed Mahnken, A.H., Spreafico, C., Maleux, G. et al.
August 4, 2014]. (2013). Standards of practice in transarte-
Larson, S.M., Erdi, Y., Akhurst, T. et al. (1999). rial radioembolization. Cardiovasc Intervent
Tumor treatment response based on visual Radiol 36:613–622. Available at http://
and quantitative changes in global tumor www.ncbi.nlm.nih.gov/pubmed/23511991
glycolysis using PET-FDG imaging. The visual [Accessed August 4, 2014].
response score and the change in total lesion Majno, P., Mentha, G., Toso, C. et al. (2014).
glycolysis. Clin Positron Imaging 2:159–171. Anatomy of the liver: An outline with three
Available at http://www.ncbi.nlm.nih.gov/ levels of complexity–A further step toward
pubmed/14516540. tailored territorial liver resections. J Hepatol
Lau, W.Y., Leung, W.T., Ho, S. et al. (1994). 60:654–662. Available at http://www.ncbi.
Treatment of inoperable hepatocellular car- nlm.nih.gov/pubmed/24211738 [Accessed
cinoma with intrahepatic arterial yttrium-90 October 13, 2014].
microspheres: A phase I and II study. Br J Mazzaferro, V., Sposito, C., Bhoori, S. et al.
Cancer 70:994–999. (2013). Yttrium-90 radioembolization for
Lausch, A., Sinclair, K., Lock, M. et al. (2013) intermediate-advanced hepatocellular
Determination and comparison of radio- carcinoma: A phase 2 study. Hepatology
therapy dose responses for hepatocel- 57:1826–1837.
lular carcinoma and metastatic colorectal Morshedi, M.M., Bauman, M., Rose, S.C.,
liver tumours. Br J Radiol 86:20130147. Kikolski, S.G. (2014). Yttrium-90 resin micro-
Available at http://www.ncbi.nlm.nih.gov/ sphere radioembolization using an antire-
pubmed/23690438. flux catheter: An alternative to traditional
Lencioni, R., Llovet, J.M. (2010). Modified coil embolization for nontarget protection.
RECIST (mRECIST) assessment for hepatocel- Cardiovasc Intervent Radiol 38:381–388.
lular carcinoma. Semin Liver Dis 30:52–60. Available at http://www.ncbi.nlm.nih.gov/
Available at http://www.ncbi.nlm.nih.gov/ pubmed/24989143 [Accessed July 18, 2014].
pubmed/20175033. Mouli, S., Memon, K., Baker, T. et al. (2013).
Leung, T.W., Lau, W.Y., Ho, S.K. et al. (1995). Yttrium-90 radioembolization for intrahepatic
Radiation pneumonitis after selective internal cholangiocarcinoma: Safety, response, and
radiation treatment with intraarterial 90yttrium- survival analysis. J Vasc Interv Radiol 24:1227–
microspheres for inoperable hepatic tumors. Int 1234. Available at http://www.pubmedcentral.
J Radiat Oncol Biol Phys 33:919–924. nih.gov/articlerender.fcgi?artid=3800023&too
Lewandowski, R.J., Sato, K.T., Atassi, B. et al. l=pmcentrez&rendertype=abstract [Accessed
(2007). Radioembolization with 90Y micro- August 4, 2014].
spheres: Angiographic and technical con- Muller, J.H., Rossier, P.H. (1951). A new
siderations. Cardiovasc Intervent Radiol method for the treatment of cancer of
30:571–592. Available at http://www.ncbi. the lungs by means of artificial radioac-
nlm.nih.gov/pubmed/17516113 [Accessed tivity. Acta Radiol 35:449–468. Available
August 4, 2014]. at http://www.tandfonline.com/doi/
Liu, D.M., Salem, R., Bui, J.T. et al. (2005). full/10.3109/00016925109136677.
Angiographic considerations in patients Nijsen, J.F.W., Van Steenbergen, M.J.,
undergoing liver-directed therapy. J Vasc Kooijman, H. et al. (2001). Characterization
Interv Radiol 16:911–935. Available at http:// of poly(L-lactic acid) microspheres loaded
www.ncbi.nlm.nih.gov/pubmed/16002500 with holmium acetylacetonate. Biomaterials
[Accessed August 4, 2014]. 22:3073–3081.
Llovet, J.M., Di Bisceglie, A.M., Bruix, J. et al. Padia, S.A., Alessio, A., Kwan, S.W. et al. (2013).
(2008). Design and endpoints of clinical Comparison of positron emission tomogra-
trials in hepatocellular carcinoma. J Natl phy and bremsstrahlung imaging to detect
28 Introduction to hepatic radioembolization

particle distribution in patients undergo- radioembolization. Int J Radiat Oncol Biol


ing yttrium-90 radioembolization for large Phys 79:163–171. Available at http://www.ncbi.
hepatocellular carcinomas or associated nlm.nih.gov/pubmed/20421150 [Accessed
portal vein thrombosis. J Vasc Interv Radiol August 4, 2014].
24:1147–1153. Available at http://www.ncbi. Riaz, A., Lewandowski, R.J., Kulik, L.M. et al.
nlm.nih.gov/pubmed/23792126 [Accessed (2009). Complications following radioem-
August 4, 2014]. bolization with yttrium-90 microspheres:
Pasciak, A.S., Bourgeois, A.C., McKinney, J.M. A comprehensive literature review. J Vasc
et al. (2014). Radioembolization and the Interv Radiol 20:1121–1130; quiz 1131.
dynamic role of (90)Y PET/CT. Front Oncol Available at http://www.ncbi.nlm.nih.gov/
4:38. Available at http://www.pubmedcentral. pubmed/19640737 [Accessed July 16, 2014].
nih.gov/articlerender.fcgi?artid=3936249&too Rosenbaum, C.E.N.M., van den Bosch, M.A.A.J.,
l=pmcentrez&rendertype=abstract [Accessed Veldhuis, W.B. et al. (2013). Added value of
August 4, 2014]. FDG-PET imaging in the diagnostic workup
Pasciak, A.S., Mcelmurray, J.H., Bourgeois, for yttrium-90 radioembolisation in patients
A.C. et al. (2015). The impact of an antire- with colorectal cancer liver metastases. Eur
flux catheter on target volume particulate Radiol 23:931–937. Available at http://www.
distribution in liver-directed embolotherapy: ncbi.nlm.nih.gov/pubmed/23111818 [Accessed
A pilot study. J Vasc Interv Radiol 26:660–669. August 4, 2014].
Available at http://www.ncbi.nlm.nih.gov/ Rosler, H., Triller, J., Baer, H.U. et al. (1994).
pubmed/25801854. Superselective radioembolization of hepato-
Prince, J.F., Smits, M.L.J., Krijger, G.C. et al. cellular carcinoma: 5-year results of a pro-
(2014a). Radiation emission from patients spective study. Nuklearmedizin 33:206–214.
treated with holmium-166 radioemboliza- Salem, R., Lewandowski, R.J., Sato, K.T. et al.
tion. J Vasc Interv Radiol 25:1956–1963.e1. (2007). Technical aspects of radioemboliza-
Available at http://www.ncbi.nlm.nih.gov/ tion with 90Y microspheres. Tech Vasc Interv
pubmed/25311966 [Accessed January 8, Radiol 10:12–29. Available at http://www.ncbi.
2015]. nlm.nih.gov/pubmed/17980315 [Accessed
Prince, J.F., van den Hoven, A.F., van den Bosch, August 4, 2014].
M.A.A.J. et al. (2014b). Radiation-induced Samuelson, S.D., Louie, J.D., Sze, D.Y. (2013).
cholecystitis after hepatic radioembolization: N-butyl cyanoacrylate glue embolization of
Do we need to take precautionary measures? arterial networks to facilitate hepatic arterial
J Vasc Interv Radiol 25:1717–1723. Available skeletonization before radioembolization.
at http://linkinghub.elsevier.com/retrieve/pii/ Cardiovasc Intervent Radiol 36:690–698.
S1051044314006447. Available at http://www.ncbi.nlm.nih.gov/
Prince, J.F., van Rooij, R., Bol, G.H. et al. (2015). pubmed/23070102.
Safety of a scout dose preceding hepatic Sharma, H. (2014). Role of external beam radia-
radioembolization with 166Ho microspheres. tion therapy in management of hepatocellular
J Nucl Med 56:817–823. Available at http:// carcinoma. J Clin Exp Hepatol 4:S122–S125.
jnm.snmjournals.org/cgi/doi/10.2967/ Available at http://linkinghub.elsevier.com/
jnumed.115.155564. retrieve/pii/S0973688314002989.
Prinzmetal, M., Ornitz, E.M. (1948). Arterio- Smits, M.L.J., Elschot, M., Sze, D.Y. et al.
venous anastomoses in liver, spleen, and (2014). Radioembolization dosimetry: The
lungs. Am J Physiol 152:48–52. road ahead. Cardiovasc Intervent Radiol.
Rengo, M., Bellini, D., De Cecco, C.N. et al. Available at http://www.ncbi.nlm.nih.gov/
(2011). The optimal contrast media policy in pubmed/25537310 [Accessed December 30,
CT of the liver. Part II: Clinical protocols. Acta 2014].
Radiol 52:473–480. Smits, M.L.J., Elschot, M., van den Bosch,
Riaz, A., Gates, V.L., Atassi, B. et al. (2011). M.A.A.J. et al. (2013a). In vivo dosim-
Radiation segmentectomy: A novel etry based on SPECT and MR imaging of
approach to increase safety and efficacy of 166Ho-microspheres for treatment of liver
References 29

malignancies. J Nucl Med 54:2093–2100. van den Hoven, A.F., Lam, M.G.E.H., Jernigan, S.
Available at http://www.ncbi.nlm.nih.gov/ et al. (2015a). Innovation in catheter design
pubmed/24136931 [Accessed August 4, 2014]. for intra-arterial liver cancer treatments
Smits, M.L.J., van den Hoven, A.F., Rosenbaum, results in favorable particle-fluid dynamics.
C.E.N.M. et al. (2013b). Clinical and labora- J Exp Clin Cancer Res 34:74. Available at
tory toxicity after intra-arterial radioemboliza- http://www.jeccr.com/content/34/1/74.
tion with 90Y-microspheres for unresectable van den Hoven, A.F., Prince, J.F., de Keizer, B. et al.
liver metastases. PLoS One 8:e69448. (2015b). Use of C-Arm Cone beam CT during
Available at http://journals.plos.org/plosone/ hepatic radioembolization: Protocol optimiza-
article?id=10.1371/journal.pone.0069448. tion for extrahepatic shunting and parenchymal
Srinivas, S.M., Natarajan, N., Kuroiwa, J. et al. enhancement. Cardiovasc Intervent Radiol.
(2014). Determination of radiation absorbed Available at http://link.springer.com/10.1007/
dose to primary liver tumors and normal s00270-015-1146–1148.
liver tissue using post-radioembolization van den Hoven, A.F., Prince, J.F., Samim,
90Y PET. Front Oncol 4:1–12. Available at M. et al. (2014a). Posttreatment PET-CT-
http://journal.frontiersin.org/journal/10.3389/ confirmed intrahepatic radioembolization
fonc.2014.00255/full. performed without coil embolization, by
Strigari, L., Sciuto, R., Rea, S. et al. (2010). Efficacy using the antireflux surefire infusion system.
and toxicity related to treatment of hepa- Cardiovasc Intervent Radiol 37:523–528.
tocellular carcinoma with 90Y-SIR spheres: Available at http://www.ncbi.nlm.nih.gov/
Radiobiologic considerations. J Nucl Med pubmed/23756882 [Accessed August 5,
51:1377–1385. 2014].
Tirkes, T., Hollar, M.A., Tann, M. et al. (2013). van den Hoven, A.F., Smits, M.L.J., de Keizer, B.
Response criteria in oncologic imaging: et al. (2014b). Identifying aberrant hepatic
Review of traditional and new criteria. arteries prior to intra-arterial radioem-
Radiographics 33:1323–1341 Available at bolization. Cardiovasc Intervent Radiol.
http://eutils.ncbi.nlm.nih.gov/entrez/eutils/ Available at http://www.ncbi.nlm.nih.gov/
elink.fcgi?dbfrom=pubmed&id=2402592 pubmed/24469409 [Accessed August 5,
7&retmode=ref&cmd=prlinks\npapers2:// 2014].
publication/doi/10.1148/rg.335125214. van den Hoven, A.F., van Leeuwen, M.S., Lam,
Tsurusaki, M., Okada, M., Kuroda, H. et al. (2014). M.G.E.H., van den Bosch, M.A.A.J. (2015).
Clinical application of 18F-fluorodeoxyglucose Hepatic arterial configuration in relation to
positron emission tomography for assessment the segmental anatomy of the liver; observa-
and evaluation after therapy for malignant tions on MDCT and DSA relevant to radio-
hepatic tumor. J Gastroenterol 49:46–56. embolization treatment. Cardiovasc Intervent
Available at http://link.springer.com/10.1007/ Radiol: 38:100–111. Available at http://www.
s00535-013-0790-5. ncbi.nlm.nih.gov/pubmed/24603968.
van de Maat, G.H., Seevinck, P.R., Elschot, M. van Leeuwen, M.S., Fernandez, M.A., van, Es.H.W.
et al. (2013). MRI-based biodistribution et al. (1994). Variations in venous and segmen-
assessment of holmium-166 poly(L-lactic tal anatomy of the liver: Two- and three-dimen-
acid) microspheres after radioembolisation. sional MR imaging in healthy volunteers. AJR
Eur Radiol 23:827–835. Available at http:// Am J Roentgenol 162:1337–1345. Available at
www.pubmedcentral.nih.gov/articlerender.fcg http://www.ncbi.nlm.nih.gov/pubmed/8191995
i?artid=3563959&tool=pmcentrez&rendertyp [Accessed August 5, 2014].
e=abstract [Accessed August 4, 2014]. Vouche, M., Lewandowski, R.J., Atassi, R. et
Van de Wiele, C., Maes, A., Brugman, E. et al. al. (2013). Radiation lobectomy: Time-
(2012). SIRT of liver metastases: Physiological dependent analysis of future liver remnant
and pathophysiological considerations. volume in unresectable liver cancer as a
Eur J Nucl Med Mol Imaging 39:1646–1655. bridge to resection. J Hepatol 59:1029–1036.
Available at http://www.ncbi.nlm.nih.gov/ Available at http://www.ncbi.nlm.nih.gov/
pubmed/22801733 [Accessed July 16, 2014]. pubmed/23811303. Accessed August 4, 2014.
30 Introduction to hepatic radioembolization

Walker, L.A. (1964). Radioactive yttrium 90: A Xing, M., Prajapati, H.J., Dhanasekaran, R. et al.
review of its properties, biological behavior, (2014). Selective internal yttrium-90 radio-
and xlinical uses. Acta Radiol Ther Phys Biol embolization therapy (90Y-SIRT) versus best
2:302–314. supportive care in patients with unresectable
Wang, S., He, X., Li, Z. et al. (2010). metastatic melanoma to the liver refractory
Characterization of the middle hepatic to systemic therapy. Am J Clin Oncol 00:1.
artery and its relevance to living donor liver Available at http://www.ncbi.nlm.nih.gov/
transplantation. Liver Transplant 16:736–741. pubmed/25089529.
Available at http://onlinelibrary.wiley.com/ Yu, N., Srinivas, S.M., Difilippo, F.P. et al. (2013).
doi/10.1002/lt.22082/abstract. Lung dose calculation with SPECT/CT for
90
Wollner, I., Knutsen, C., Smith, P. et al. (1988). yittrium radioembolization of liver cancer.
Effects of hepatic arterial yttrium 90 glass Int J Radiat Oncol Biol Phys 85:834–839.
microspheres in dogs. Cancer 61:1336–1344. Available at http://www.ncbi.nlm.nih.gov/
Wondergem, M., Smits, M.L.J., Elschot, M. pubmed/22871239.
et al. (2013). 99mTc-macroaggregated Zielhuis, S.W., Nijsen, J.F.W., de Roos, R. et al.
albumin poorly predicts the intrahepatic (2006). Production of GMP-grade radioactive
distribution of 90Y resin microspheres in holmium loaded poly(L-lactic acid) micro-
hepatic radioembolization. J Nucl Med spheres for clinical application. Int J Pharm
54:1294–1301. Available at http://www.ncbi. 311:69–74. Available at http://www.ncbi.nlm.
nlm.nih.gov/pubmed/23749996 [Accessed nih.gov/pubmed/16439073 [Accessed August
July 10, 2014]. 4, 2014].
PART 2
Patient Selection and Treatment
Planning

2 Treatment options for patients with primary and secondary liver cancer: An overview
of invasive, minimally invasive, and noninvasive techniques 33
Ricardo Paz-Fumagalli, David M. Sella, and Gregory T. Frey
3 Treatment planning part I: Vascular considerations associated with safety and efficacy
in radioembolization 53
Ray Bradford and J. Mark McKinney
4 Treatment planning part II: Procedure simulation and prognostication 65
Shyam M. Srinivas, Sankaran Shrikanthan, Naichang Yu, Susan D. Kost, Ram Gurajala, and
Karunakaravel Karuppasamy
5 Treatment planning part III: Dosimetric considerations in radioembolization with glass
and resin microspheres 87
Alexander S. Pasciak, Austin C. Bourgeois, and Yong C. Bradley
6 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy 113
Edward Kim, Joseph Titano, and Safet Lekperic
2
Treatment options for patients with
primary and secondary liver cancer:
An overview of invasive, minimally
invasive, and noninvasive techniques

RICARDO PAZ-FUMAGALLI, DAVID M. SELLA, AND GREGORY T. FREY

2.1 Introduction 33 2.6 Colorectal liver metastases 44


2.2 Approaches to the patient with 2.6.1 Medical therapy for CRC 44
hepatic malignancy 34 2.6.2 Surgical therapy for CRC 44
2.3 Systemic therapies 34 2.6.3 Chemoembolization for CRC 44
2.4 Locoregional therapies 35 2.6.4 Radioembolization 44
2.4.1 Surgery 35 2.6.5 Tumor ablation for CRC 45
2.4.2 Tumor ablation 36 2.7 Metastatic NET 45
2.4.3 Transarterial therapy 38 2.7.1 Surgery for NET 45
2.5 Hepatocellular cancer 42 2.7.2 Tumor ablation for NET 46
2.5.1 Surgical resection and liver 2.7.3 Systemic therapy for NET 46
transplantation for HCC 43 2.7.4 Transarterial therapy for NET 46
2.5.2 Tumor ablation for HCC 43 2.8 Intrahepatic cholangiocarcinoma 47
2.5.3 Chemoembolization for HCC 43 References 47
2.5.4 Radioembolization for HCC 43
2.5.5 Systemic therapy for HCC 43

of liver cancer are generally determined by the


2.1 INtrODUCtION tumor size, number, location, proximity to vul-
nerable structures, rate of growth, production of
Liver cancer occurs when cellular proliferation substances, pattern of spread, and underlying con-
within the liver escapes normal control mecha- dition of the patient’s organ systems. Oncologists
nisms, exhibits aggressive behavior, and reaches study cancer populations rather than individu-
or invades other body parts causing deterioration als and express outcomes in statistical terms that
of the patient’s well-being and shortening of the reflect cancer control or freedom from cancer
patient’s life expectancy. It may start in the liver (overall survival, median survival, cancer-specific
(primary) or reach the liver from another origin survival, disease-free survival, progression-free
(secondary or metastatic). The signs and symptoms survival, time-to-progression) or can reflect

33
34 Treatment options for patients with primary and secondary liver cancer

improvement, maintenance, or deterioration of liver failure (Lewandowski and Davenport, 2015).


quality of life. Various models and staging systems can be uti-
Cancer treatment aims to maintain or improve lized to predict overall performance status, dis-
quality of life and lifespan and can be extremely ease burden, hepatic reserve, and prognosis. The
complex due to the wide variety of tumor types, model for end-stage liver disease (MELD) and
the presence or absence of underlying liver disease, Child–Pugh scores take into account various
and a multitude of available therapies. Treatment chemical and clinical factors including creatinine,
can be systemic (whole body) or locoregional total bilirubin, serum albumin, prothrombin time,
(organ- or volume-specific) and can have a cura- ascites, and encephalopathy to assess progno-
tive intent or be meant to lessen the impact of the sis of chronic liver disease/cirrhosis. The Eastern
disease (palliation). Treatment objectives can cross Cooperative Oncology Group (ECOG) scale of
over; occasionally, a treatment given with pallia- performance status is used for measuring how a
tive intent can later enable a curative approach. disease impacts a patient’s daily living abilities. It
This book is focused specifically on one form determines a patient’s level of functioning in terms
of liver cancer treatment: yttrium-90 (90Y) radio- of daily activity, physical ability, and self-care abil-
embolization. However, because of the multidis- ity. Finally, one must consider the goals of therapy.
ciplinary nature of radioembolization, many of When designing a treatment plan, one must take
the individuals involved in a radioembolization into consideration factors such as the extent of dis-
program may not be very familiar with other treat- ease, both intra- and extrahepatic, as well as previ-
ments for hepatic malignancy. Those who directly ous therapies. The treatment of liver malignancies
or indirectly contribute to the care of a patient can be complex, and a multispecialty team may
receiving 90Y radioembolization should have a include medical oncologists, hepatologists, sur-
basic understanding of the therapies a patient has geons, radiologists, and radiation oncologists.
received or may receive in the future. To frame the
context in which the role of radioembolization can
be appreciated, this chapter provides an overview
of standard-of-care treatment modalities for pri- 2.3 SYStEMIC tHEraPIES
mary and secondary liver cancer and discusses
the strengths, weaknesses, and contraindications Systemic therapies act throughout the body. The
of the therapeutic options stratified by tumor type. most important categories include cytotoxic che-
motherapy, hormonal therapy, and targeted therapy.
Cytotoxic chemotherapy interferes with the
different steps in the cell cycle and directly tar-
2.2 aPPrOaCHES tO tHE
gets and kills cancer cells but it is also harmful to
PatIENt WItH HEPatIC
normal cells. There are numerous cytotoxic phar-
MaLIGNaNCY macologic mechanisms and agents. Alkylating
agents interact with DNA (cyclophosphamide,
When approaching the patient with hepatic can- cisplatin, oxaliplatin), antimetabolites interfere
cer, one must first determine whether the process with DNA precursors and cellular metabolism
is primary or metastatic. Hepatocellular carci- (5-fluorouracil, gemcitabine), and antitumor
noma (HCC) is the most common primary liver antibiotics interfere with DNA activity (mito-
tumor (Ferlay et al., 2010). Colorectal cancer is a mycin, doxorubicin). Topoisomerase inhibitors
common secondary tumor of the liver. Other liver (irinotecan) and mitotic inhibitors (vincristine,
malignancies commonly treated with locoregional paclitaxel, docetaxel) represent other mecha-
therapies include neuroendocrine tumor (NET) nisms. Cytotoxic chemotherapy is limited by its
metastases, breast cancer metastases, and intra- nonspecificity, as it is toxic to both cancer and
hepatic cholangiocarcinoma (ICC). Second, the normal cells, resulting in the adverse effects and
synthetic function of the patient’s liver should be toxicities observed during therapy.
considered. Preserved liver function is important Hormonal therapy is a broad category that
because the antitumor effects of various thera- includes inhibitors of hormone synthesis (letro-
pies can be counteracted by treatment-induced zole), hormone receptor antagonists (tamoxifen),
2.4 Locoregional therapies / 2.4.1 Surgery 35

and hormone supplements (estrogens, proges- 2.4.1 SURGERY


tins, androgens, and somatostatin analogs such as
octreotide). Hormonal therapy has an anticancer The objective of liver surgery is to remove the malig-
effect but is also used to alleviate the symptoms of nant tissue surrounded by a margin of tumor-free
hormone- and peptide-secreting tumors. liver (resection) or the entire liver (transplantation).
Targeted therapy promises to minimize the The success and safety are determined by proper
problems that cytotoxic chemotherapy causes patient selection based on the patient’s health, tumor
because it interferes with specific molecules that characteristics, the anatomy of liver in relation to
participate in the genesis of cancer and tumor the malignancy, the quality and volume of the liver
growth rather than interfering nonspecifically in remnant after the resection is complete, and adher-
cell multiplication, which is a function shared with ence to established criteria for transplantation.
normal tissues. These agents are often called bio- The type of resection is chosen after determining
logicals, and the most commonly used in liver can- that the future liver remnant is large enough (usually
cer include tyrosine kinase inhibitors (sorafenib, >30% of total liver volume), that the liver functional
sunitinib, erlotinib, imatinib), serine/threonine reserve is adequate, and that there is the possibility of
kinase inhibitors (everolimus), and monoclonal achieving a surgical margin negative for residual can-
antibodies [bevacizumab has an antiangiogenesis cer. The surgery can follow the segmental anatomy of
effect by blocking vascular endothelial growth fac- the liver and varies in extent from segmentectomy, to
tor A (VEGF-A), and cetuximab inhibits epidermal lobectomy, and to extended hepatectomy. The resec-
growth factor receptor (EGFR)]. Immunotherapy is tion can also be nonanatomic, which is often referred
a variant of targeted therapy that has the objective to as wedge resection (Kishi et al., 2009).
of activating the immune system to identify and If the expected liver remnant is too small, it
reject the cancer cells. Monoclonal antibody-based is possible to induce growth of the remnant by
therapy is the most common and successful form interrupting portal vein perfusion of the liver
of cancer immunotherapy. Cytokines (interleukin, portion to be removed. Preoperative portal vein
interferon) and cellular therapy (cancer vaccines) embolization (PVE) and the so-called “associ-
are other forms of immunotherapy. ating liver partition and portal vein ligation for
staged hepatectomy” (ALPPS) surgical procedure
are the two procedures designed to achieve this
effect (Shindoh et al., 2013a, 2013b). The physi-
2.4 LOCOrEGIONaL tHEraPIES ologic response in the liver remnant is called the
atrophy–hypertrophy complex (Kim et al., 2008).
Locoregional therapy of liver cancer is directed to PVE is performed by interventional radiologists
a well-defined zone and lacks any activity outside using multiple techniques to achieve complete
of the volume defined by the treatment plan. The occlusion of the portal vein branches expected to
therapy can be surgical, minimally invasive guided be removed (Guiu et al., 2013). PVE is safe and
by imaging, or noninvasive in the form of exter- has a low complication rate (Abulkhir et al., 2008;
nal radiation therapy. Surgical options include Ratti et al., 2010). However, questions have been
tumor resection, intraoperative thermal ablation, raised about the potential of tumor growth fol-
and liver transplantation. Image-guided therapy lowing embolization related to alterations in blood
is most commonly done percutaneously by means flow patterns and increases in local growth factors
of needles or probes inserted into the body and (Simoneau et al., 2012). Another method to induce
directed with computed tomography (CT), ultra- hypertrophy of the functional liver remnant is
sound, magnetic resonance imaging (MRI), or flu- lobar radioembolization with Y-90 microspheres
oroscopy, or it can be administered into the hepatic (Fernandez-Ros et al., 2014; Garlipp et al., 2014;
arterial blood flow after catheterization. Radiation Teo and Goh, 2015).
therapy for liver tumors is most commonly given Liver transplantation is well established as the
in tightly focused external beams that use stereo- treatment of choice for HCC, and HCC accounts
tactic techniques or delivered during catheteriza- for the vast majority of transplantation for malig-
tion into the hepatic arterial circulation to achieve nancy, but metastatic NETs, epithelioid heman-
selective internal radiation therapy. gioendothelioma, and cholangiocarcinoma are
36 Treatment options for patients with primary and secondary liver cancer

occasionally treated with transplantation. Liver that heals into a scar over time (Figure 2.1). Tissue
transplantation is particularly valuable for HCC destruction can be achieved with extreme tem-
because it simultaneously treats the hepatic malig- perature by inducing heating or freezing (radiofre-
nancy and the underlying cirrhosis and/or liver quency ablation or RFA, laser ablation, microwave
insufficiency (Eghtesad and Aucejo, 2014). ablation [MW], high-intensity focused ultrasound,
cryoablation), with injection of chemicals such as
2.4.2 TUMOR ABLATION alcohol and concentrated acetic acid and by dis-
rupting the cell membrane at the molecular level
Tumor ablation intends to destroy tissue in the with electrical fields (irreversible electroporation
body without removal, leaving a zone of necrosis or IRE) (Ahmed, 2014). For percutaneous ablation,

(a) (b)

(c)

Figure 2.1 MR images after intravenous contrast processed with subtraction technique in a
67-year-old man with hepatocellular carcinoma. (a) Single intensely enhancing hepatocellular car-
cinoma in segment VII of the liver (arrowheads). (b) One month after percutaneous radiofrequency
ablation, the zone of ablation is sharply defined without any residual abnormal contrast enhancement
to indicate residual tumor (arrowheads). (c) At 76 years of age, 9 years after the ablation, the treated
volume has decreased (arrowheads) and there is no abnormal enhancement to indicate active tumor.
2.4 Locoregional therapies / 2.4.2 Tumor ablation 37

probes (thermal ablation, IRE) or needles (chemi- 2450 MHz. The probe is tuned to the natural fre-
cal injection) are inserted through skin punctures quency of water, and similarly to RFA, vigorous
and directed to the treatment zone guided with motion occurs at the molecular level during appli-
imaging, typically CT or ultrasound, but other cation of microwaves, producing frictional heat.
imaging methods can be used. Tumor ablation can MW has several advantages over RFA. It is faster,
also be guided surgically, whether laparoscopically can achieve higher temperatures, is less suscep-
or through a laparotomy. The number and distri- tible to heat-sink effect, and does not depend on
bution of the probes or needles will define a spe- electrical conductivity so it is not encumbered by
cific kill volume. Tumor ablation can be performed poorly conducting tissues such as bone or air-filled
alone or in combination with other treatments lung. The devices are capable of operating multiple
including chemotherapy, surgery, or embolization probes simultaneously. Large ablation volumes
(Wells et al., 2015). Obesity, under-lying liver dis- ≥5 cm can be reached more reliably than with RFA
ease, and tumor size and location can affect both the (Simon et al., 2005).
results and frequency of complications with tumor
ablation (Komorizono et al., 2003; Livraghi et al., 2.4.2.3 Cryoablation
2003; Teratani et al., 2006).
The clinically available cryoablation probes are
2.4.2.1 Radiofrequency ablation designed to circulate a gas, such as argon, into an
expansion chamber at the tip of the probe, which
Radiofrequency ablation applies to target tissues causes profound cooling (Joule–Thompson effect).
alternating current at a high frequency, similar to The adjacent tissues freeze and the frozen volume
electrocautery. The term radiofrequency is mislead- expands over time. Because cells suffer harm both
ing because this method does not apply electro- from freezing as well as thawing, a commonly uti-
magnetic energy in the form of radio waves; rather, lized sequence requires freezing for 10 min, thaw-
it refers to the frequency of the alternating current, ing for 8 min, and refreezing for 10 min. Tissue is
typically 460–500 kHz, that falls within the fre- reliably devitalized with this type of protocol at
quency range of radio. As the current alternates, temperatures below –20°C. Therefore, frozen tis-
dipolar molecules and ions such as sodium and sues at the edges of the ice ball survive, and the kill
potassium move quickly to align with the current, zone is smaller than the size of the freeze, as shown
inducing frictional heat. Temperatures can rise with intraprocedural CT.
well above the boiling point of water. Temperatures The x-ray beam attenuation of ice is less than
>50°C are quickly lethal at the cellular level. water, making the frozen volume very clearly vis-
The probes function as electrodes in the electri- ible with CT. Cryoablation is, therefore, advanta-
cal circuit that runs through the targeted tissues. geous when sharp visibility of the ablation edge
The size of the ablation volume and extent of cell is needed to monitor and protect nearby vulner-
death with RFA are limited by the electrical con- able structures from injury. Multiple probes used
ductivity of the tissue, patterns of heat convection simultaneously permit sculpting larger volumes of
and conduction, and the presence of “heat sinks.” ablation than are achievable with heat-based sys-
Flowing blood in medium to large vessels can cool tems. Cryoablation causes less pain that RFA or
nearby tissues during RFA and prevent reaching MW and is less destructive of the connective tissue
lethal temperatures. RFA is relatively slow and is structure. For these reasons, it is better suited for
unreliable for ablation volumes with diameters >5 locations where substantial postprocedural pain
cm; however, it is quite reliable for volumes ≤3 cm can be expected and when attempting to preserve
(Hong and Georgiades, 2010). Of all thermal abla- the integrity of an adjacent structure.
tion methods, RFA is the most studied and most Because cryotherapy does not have a cauteriz-
widely reported in the medical literature. ing effect like RFA or MW and the probe diam-
eter tends to be larger, there is greater potential
2.4.2.2 Microwave ablation for postprocedural hemorrhage, which limits the
application of percutaneous cryotherapy for liver
MW probes are antennae that broadcast elec- tumors. Caution must be exercised when creating
tromagnetic waves with frequencies from 900 to large ice balls because of the threat of cryoshock,
38 Treatment options for patients with primary and secondary liver cancer

a severe consequence of therapy that can be fatal, 2.4.3.2 Chemoembolization


and because of the threat of tissue fracture with
hemorrhage (Erinjeri and Clark, 2010). Transcatheter arterial chemoembolization (TACE)
treats liver cancer with a combination of local
2.4.2.4 Irreversible electroporation delivery of tumorcidal chemicals in a highly con-
centrated form while inducing tumor ischemia
Irreversible electroporation units apply high- and maintaining very low concentrations of sys-
voltage current across a cellular membrane, which temic chemotherapeutic agents (Lencioni, 2010).
creates small pores, destabilizes the cellular mem- The conventional TACE (cTACE) mixture is based
brane, and causes cell death (Gehl, 2003). IRE on ethiodized oil used either alone, in an emulsion
requires general anesthesia and complete muscle with chemotherapeutic agents or in combination
relaxation. Because it does not have a significant with absolute ethanol (Figure 2.3). Doxorubicin is
effect on connective tissues, it is the least likely of the most commonly used chemotherapy agent in
the ablation methods to damage vessels, bile ducts, the United States; however, epirubicin, cisplatin,
the gallbladder, or the bowel (Silk et al., 2014). and mitomycin C are also utilized. The mixture
Patients who benefit the most from IRE have is delivered through an arterial catheter placed as
limited disease in the central portion of the liver selectively as possible into the arteries feeding the
where resection and other ablative modalities can- tumor, usually followed by embolization particles
not be done safely (Lencioni et al., 2015; Scheffer that induce ischemic necrosis of the tumor and
et al., 2015). Experience with IRE at this time is prevent washout of the drug.
preliminary, and there are scant data focused on Drug-eluting bead TACE (DEB-TACE) involves
outcomes. the delivery of microspheres loaded with chemo-
therapy and provides a sustained release of drug
2.4.3 TRANSARTERIAL THERAPY and tumor vessel occlusion following intra-arterial
administration (Figure 2.4). The two agents uti-
The hepatic arteries provide another route for lized most often are doxorubicin for HCC or virtu-
locoregional therapy. Both primary and meta- ally any other tumor type drug-eluting beads with
static liver cancers derive blood supply almost doxorubicin (DEB-DOX) or drug-eluting beads
completely from the hepatic arterial circulation. with irinotecan for colorectal metastases (DEB-
In contrast, the functional liver tissue receives IRI). DEB-TACE utilizes catheter-based tech-
60%–75% of blood flow from the portal vein. niques similar to cTACE.
Transarterial therapies exploit this differential After chemoembolization, most patients experi-
perfusion. Consequently, the tumor receives a ence a postembolization syndrome of right upper
highly concentrated therapeutic dose while, for the quadrant pain, nausea, fever, and loss of appetite
most part, sparing the functional liver tissue. The that is self-limited over a period of several days.
most common transarterial options include inert Fatigue often occurs, can be profound, and can last
particle embolization, chemoembolization, and a few weeks. Reported complications include bile
radioembolization. duct injury, liver abscess (particularly after biliary
intervention), tumor rupture, and nontarget injury
2.4.3.1 Inert particle embolization with necrosis in vascular territories supplying the
bowel, gallbladder, and diaphragm. Some studies
Administration of small particles (usually 45–750 have demonstrated fewer adverse events and an
μm in size) into the hepatic arteries causes micro- improved pharmacokinetic profile with DEB-TACE
vascular occlusion and interrupts the blood sup- compared with cTACE (Liapi and Geschwind, 2010;
ply. The malignant disease quickly develops large Molvar and Lewandowski, 2015).
zones of ischemia and coagulative necrosis (Figure
2.2). Particle embolization can be very effective for 2.4.3.3 Radioembolization
treatment of HCC and metastatic NET but is less
effective with cholangiocarcinoma and metastatic The radioembolization procedure will be presented
disease of other histologic types (Brown et al., in detail elsewhere in this text. Briefly, radioem-
1999; Maluccio et al., 2008). bolization refers to the delivery of glass or resin
2.4 Locoregional therapies / 2.4.3 Transarterial therapy 39

(a) (b)

(c)

Figure 2.2 A 70-year-old man with an intestinal carcinoid tumor discovered 20 years earlier compli-
cated by liver metastases and symptoms of flushing and diarrhea partially controlled with octreotide
therapy. (a) Computed tomography (CT) scan with intravenous contrast shows multiple bulky metas-
tases in the liver with only modest enhancement. (b) Hepatic arteriography showed distortion of the
hepatic artery branches and only subtle enhancement of the disease. (c) CT scan follow-up after 6
months confirmed complete loss of the modest enhancement in most lesions and marked decrease in
the size of the tumors.

microspheres loaded with Y-90 radioactive isotope radioembolization is similar to that of chemoem-
into the liver circulation by means of catheteriza- bolization. Contraindications include established
tion. Like TACE, it takes advantage of the differen- liver insufficiency, unsuitable anatomy that places
tial arterial flow between tumor and normal tissues the patient at risk for nontarget radiation exposure
to deliver a high dose to the malignancy with mini- to the gastrointestinal tract, and hepatopulmonary
mization of the radiation dose to the normal liver shunting estimated to exceed the maximum allow-
(Lencioni, 2010). Because hepatic artery occlusion able radiation lung doses as shown on a Tc-99m
is not necessarily the goal in radioembolization, macroaggregated albumin nuclear scan done after
the typical postembolization syndrome is usu- administration of the radionuclide into the hepatic
ally avoided or minimized. Patient eligibility for artery.
40 Treatment options for patients with primary and secondary liver cancer

(a) (b)

(c) (d)

Figure 2.3 A 56-year-old woman with liver cirrhosis complicated by hepatocellular carcinoma treated
with conventional lipiodol-based chemoembolization in preparation for liver transplant. (a) Magnetic
resonance (MR) image of the liver after intravenous contrast shows intense enhancement in the tumor
(arrow). (b) Hepatic arteriogram shows enhancement that corresponds to the tumor (arrowheads).
(c) Chemoembolization during super-selective catheterization of the artery feeding directly into the
tumor achieves dense saturation of the tumor with the therapeutic agents (arrow). Another tumor
(arrowhead) has residual lipiodol from a previous treatment, a common long-term imaging finding
after conventional chemoembolization. (d) MR image with intravenous contrast 3 months after
treatment shows complete lack of enhancement, which indicates loss of viability (arrow). Pathologic
examination of the explanted liver showed complete necrosis of the targeted tumor.

Radioembolization and chemoembolization differ if standard selection criteria are met. Serious com-
in their respective adverse events. With radioemboli- plications from nontarget radiation include gastro-
zation, fatigue is common and postembolization syn- intestinal ulceration, cholecystitis, pancreatitis, and
drome is uncommon. Radioembolization-induced radiation pneumonitis (Riaz et al., 2009; Molvar and
liver disease (REILD) is an uncommon complication Lewandowski, 2015).
2.4 Locoregional therapies / 2.4.3 Transarterial therapy 41

(a) (b)

(c) (d)

Figure 2.4 A 63-year-old man with a hepatocellular carcinoma with alpha fetoprotein (AFP) tumor
marker level of 397 ng/mL. (a) CT scan of the cirrhotic liver shows invasion of the portal vein, which
intensely enhances with intravenous contrast (arrowheads). (b) Hepatic arteriogram shows intense
enhancement of the tumor invading the portal vein that matches the CT scan findings (arrows). (c)
After drug-eluting bead chemoembolization with doxorubicin, the tumor enhancement and overall
arterial flow are decreased. (d) Two months after treatment, the tumor in the portal vein was smaller,
had lost its contrast enhancement (arrowheads), and AFP decreased to 35 ng/mL. Seven months after
treatment, the AFP level was 13 ng/mL.

2.4.3.4 External-beam radiation technological developments have broadened its


therapy applicability. Conventional radiation therapy often
requires large treatment fields in most but the smallest
External-beam radiation has traditionally played of tumors. Therefore, too much liver is exposed and
a limited role in liver tumor treatment, but recent can lead to radiation-induced liver disease (RILD),
42 Treatment options for patients with primary and secondary liver cancer

Figure 2.5 Planning radiation dosimetry before stereotactic body radiation therapy for colon can-
cer that metastasized to the liver. The tightly defined volume exposed to the radiation allows higher
dosimetry with greater safety compared with conventional external-beam radiation.

a complication that can be fatal. Three-dimensional along with the spread of hepatitis C virus (Davis et
conformal radiation therapy (3D-CRT) and stereo- al., 2010). Because imaging diagnosis of HCC can
tactic body radiation therapy (SBRT) are safer. Both be reliably made with high-quality cross-sectional
methods can deliver high-dose treatment in the imaging using multiphasic contrast-enhancement
tumorcidal range while minimizing the injury to patterns, tissue confirmation with percutaneous
surrounding tissues (Figure 2.5). An advantage of biopsy is generally reserved for lesions with atypi-
SBRT is that the high-dose treatment can be given cal imaging features (Marrero et al., 2005; Forner
in one or a few sessions (Tanguturi et al., 2014). et al., 2008; Bruix and Sherman, 2011). There is
considerable variation in the epidemiology of HCC
based on risk factors present in developing versus
2.5 HEPatOCELLULar CaNCEr developed countries, including hepatitis B and C,
alcoholic cirrhosis, nonalcoholic fatty liver disease,
HCC is the most common primary liver tumor and and nonalcoholic steatohepatitis. The Barcelona
usually occurs alongside cirrhosis (Ferlay et al., 2010). Clinic Liver Cancer (BCLC) classification is
The incidence is on the rise in the United States the most commonly used staging system in the
2.5 Hepatocellular cancer / 2.5.5 Systemic therapy for HCC 43

Western HCC population and integrates tumor between interventions when using combination
burden, liver function (Child–Pugh classification), therapy. In select situations, combined emboliza-
and performance (ECOG) status to link prognosis tion and ablation appear to improve both technical
with treatment options. This provides a framework success and local tumor progression (Takaki et al.,
to select patients who receive curative versus pal- 2009; Peng et al., 2010).
liative treatments (Llovet et al., 1999). Liver trans-
plantation, resection, and ablation are considered
2.5.3 CHEMOEMBOLIZATION FOR
curative for very early- (stage 0) and early-stage
HCC (stage A). Palliative options include TACE for
HCC
intermediate-stage HCC (stage B) and radioem- The BCLC staging system recommends cTACE for
bolization and sorafenib for advanced-stage HCC intermediate stage HCC. Conventional TACE may
(stage C). Best supportive care is utilized in termi- be used as a bridge to transplant or as a palliative
nal disease (stage D) (Reig et al., 2014). therapy for unresectable HCC. Chemoembolization
has demonstrated survival benefit over conservative
2.5.1 SURGICAL RESECTION AND treatment or best supportive care in two well-known
LIVER TRANSPLANTATION randomized controlled trials (RCTs) (Lo et al., 2002;
Llovet et al., 2002). In addition, a meta-analysis of
FOR HCC
18 RCTs demonstrated a 2-year survival benefit
Surgical resection remains a curative option for of cTACE over conservative treatment (Camma et
HCC, but wide applicability is limited because of al., 2002). The PRECISION V trial was a prospec-
the decreased hepatic reserve found in most cir- tive RCT that compared the efficacy of cTACE to
rhotic livers. Liver transplantation is the defini- DEB-TACE. DEB-TACE failed to show a response
tive therapy for HCC in the setting of cirrhosis. improvement over cTACE at 6 months; however,
However, the majority of patients never receive there was a statistically significant reduction in liver
transplantation because they do not meet trans- toxicity. Subset analysis of patients with advance
plantation criteria, because of a lack of access to disease showed an improved objective response rate
a transplant center, or because of decreased organ with DEB-TACE (Lammer et al., 2010).
availability (Kim et al., 2005).
2.5.4 RADIOEMBOLIZATION FOR
2.5.2 TUMOR ABLATION FOR HCC HCC

Image-guided tumor ablation is recommended for Radioembolization plays a large role in the treat-
patients with very early- and early-stage HCC who ment of HCC. The safety and efficacy of radioembo-
are not candidates for surgical resection according lization are demonstrated in multiple retrospective
to the BCLC criteria. The reference standard for studies and cohorts. There are no RCTs comparing
ablation of small HCC is RFA. Studies have demon- radioembolization and other locoregional therapies
strated RFA to be as effective as surgical resection for HCC. Data available suggest advantages and dis-
in very early and early HCC (Cho et al., 2010). The advantages for TACE and radioembolization, but
evidence available suggests that microwave is at least clear superiority has not been shown for either ther-
equivalent to RFA in the treatment of very early- and apy (Moreno-Luna et al., 2013; Minocha et al., 2014).
early-stage HCC (Groeschl et al., 2014; Ziemlewicz et
al., 2015; Vogl et al., 2015). RFA and MW can be con- 2.5.5 SYSTEMIC THERAPY FOR HCC
sidered first-line therapy in patients with liver dys-
function who have very early- and early-stage HCC HCC is chemotherapy resistant and systemic
in favorable locations (Wells et al., 2015). chemotherapy is generally not well tolerated in
Combination therapy harnesses the synergy patients with the significant hepatic dysfunction
between different treatment modalities. With that accompanies cirrhosis. Because survival of
HCC, this most commonly involves the use of par- patients with advanced HCC is often determined
ticle embolization or TACE with a heat-based abla- by the degree of hepatic dysfunction rather than
tion. There is no consensus on the order or timing the tumor, it is difficult to determine the benefit
44 Treatment options for patients with primary and secondary liver cancer

from chemotherapy. Sorafenib is an orally active discussed the potential for perioperative complica-
multikinase inhibitor acting on the vascular endo- tions in patients taking bevacizumab because of its
thelial growth factor receptor (VEGFR). Results effect on the vasculature (Scappaticci et al., 2005;
from the phase III SHARP (sorafenib hepatocel- Gordon et al., 2009). Arterial dissection is a well-
lular carcinoma assessment randomized protocol) known complication that can occur spontaneously
trial suggested a survival benefit compared with and increases the risk and difficulty of transarte-
best supportive care. The multicenter SHARP trial rial therapies for liver metastases that require cath-
randomly assigned 602 patients with inoperable eterization (Aragon-Ching et al., 2008; Brown,
HCC and Child–Pugh A cirrhosis to sorafenib or 2011; Mantia-Smaldone et al., 2013).
placebo. Overall survival was significantly longer in
the sorafenib-treated patients (10.7 vs. 7.9 months)
2.6.2 SURGICAL THERAPY FOR CRC
along with time to radiologic progression (5.5 vs. 2.8
months). Major adverse effects included diarrhea Surgical excision of primary and metastatic dis-
and hand–foot skin reaction. This study established ease must be considered in all patients and done
sorafenib monotherapy as the standard systemic when possible because resection provides improved
treatment for advanced HCC (Llovet et al., 2008). 5-year survival rates compared with nonoperative
candidates (Van Cutsem et al., 2006). Surgery is typ-
ically performed in conjunction with systemic che-
2.6 COLOrECtaL LIVEr motherapy. The timing of surgical management and
MEtaStaSES administration of chemotherapy can be variable
(Benson et al., 2014). PVE and radioembolization of
the portion of liver to be resected increase the num-
Colorectal cancer (CRC) is the second leading cause ber of patients eligible for surgery by increasing the
of cancer deaths in the United States (Siegel et al., volume of the functional liver remnant (Kabbinavar
2013). Of patients with CRC, approximately half et al., 2005; Abulkhir et al., 2008; Ratti et al., 2010).
develop metastases, and the liver is the most com-
mon site for these (Yoo et al., 2006). The majority
of patients with metastatic disease are unresectable 2.6.3 CHEMOEMBOLIZATION FOR
(Muratore et al., 2007). Metastatic colorectal can- CRC
cer (mCRC) is best treated by a multidisciplinary
Chemoembolization for mCRC is not currently
team of surgeons, oncologists, interventional radi-
considered first-line therapy. The use of DEB-IRI
ologists, hepatologists, and radiation oncologists.
for treatment of mCRC was reported in 2006
(Aliberti et al., 2006). One study demonstrated
2.6.1 MEDICAL THERAPY FOR CRC improved median survival for patients undergo-
Systemic chemotherapy is the mainstay of medical ing treatment with DEB-IRI versus FOLFIRI after
therapy. The primary goal is conversion of unre- failure of first-line agents (Fiorentini et al., 2012).
sectable disease to resectable disease, when possi- Timing of locoregional therapy is best managed in
ble. Multiple chemotherapy regimens are currently discussion with other stakeholders in the patient’s
recommended as first-line therapy, including management.
FOLFOX (folinic acid, f luorouracil, oxalipla-
tin), FOLFIRI (folinic acid, f luorouracil, iri- 2.6.4 RADIOEMBOLIZATION
notecan), CapeOx (capecitabine, oxaliplatin),
infusional 5-FU/LV (-f luorouracil, leucovorin) Radioembolization with Yttrium-90 labeled resin
or capecitabine, or FOLFOXIRI (folinic acid, beads is approved by the U.S. Food and Drug
f luorouracil, oxaliplatin, irinotecan) (Benson Administration (FDA) for the treatment of mCRC.
et al., 2014). Bevacizumab, a monoclonal antibody Data for the recently completed SIRFLOX trial are
that blocks VEGF, has been used in conjunction available only in abstract form. This study com-
with 5-FU/LV regiments and has shown statisti- pared Y-90 radioembolization plus FOLFOX che-
cally significant improvement in median survival motherapy versus FOLFOX chemotherapy alone as
(Kabbinavar et al., 2005). Multiple reports have first-line treatment of nonresectable liver metastases
2.7 Metastatic NET / 2.7.1 Surgery for NET 45

from colorectal cancer. It demonstrated statistically which are responsible for most symptomatology
significant improvement in median hepatic progres- and are the main cause of death.
sion-free survival and tumor response rates in the Gastroenteropancreatic NET can have differ-
group that received radiation (Gibbs et al., 2014). ent degrees of malignant behavior. Pathological
criteria of tumor grade are based on markers
2.6.5 TUMOR ABLATION FOR CRC of cell division, such as the number of mitoses
per 10 high-power fields and Ki-67 index, a cell
Long-term survival rates of patients carefully proliferation marker. Low-grade typical carci-
selected for treatment with RFA are comparable to noid shows <2 mitoses/10 high-power field (hpf)
rates of resected patients (Raut et al., 2005; Amersi and <3% Ki-67, intermediate grade atypical
et al., 2006). Local recurrence rates, however, carcinoid: 2–20 mitoses/10 hpf or 3–20% Ki-67,
remain higher (Solbiati et al., 1997). Tumor abla- and high-grade small- or large-cell neuroendo-
tion is also frequently used in combination with crine carcinoma >20 mitoses/hpf or >20% Ki-67
resection when complete resection is not feasible (Kunz, 2015).
(Ito et al., 2010). Numerous factors determine treatment.
In general, tumor ablation for mCRC is best used Management differs if the NET is limited to the
for patients with unresectable liver metastases due primary site, if the NET is metastatic, or if the pri-
to size, location, or comorbidities. RFA has 3-year mary site is unresectable. Liver-dominant metas-
survival rates of 46% and 5-year survival rates of tases, substantial extrahepatic disease, tumor
less than 20%. Ablation is also associated with volume, tumor growth rate, origin (pancreatic
higher recurrence (1 year: 12%; 5 years: 50%–70%). vs. gastrointestinal), pathologic tumor grade, and
Lesion size is a predictor of recurrence and overall whether the tumor is clinically functional are the
survival. With lesions less than 2.5 cm, the 5-year most critical factors. Pancreatic origin correlates
survival is 56%. Survival is only 13% with lesions with greater aggressiveness with median overall
greater than 2.5 cm (Boutros et al., 2010). Location survival of 24 months compared with 56 months
can also affect the rate of complications in patients for gastroenteric NET (Yao et al., 2008).
undergoing ablative therapy (Raman et al., 2004).
One study demonstrated no statistically signifi- 2.7.1 SURGERY FOR NET
cant difference in survival rates between MW and
resection (Shibata et al., 2000). Irreversible elec- When the NET is localized to its organ of origin,
troporation has been used in mCRC but the data surgery is the treatment of choice if the tumor is
are insufficient for outcomes analysis at this time deemed resectable. When the tumor is metastatic,
(Scheffer et al., 2015). surgery is still indicated if at least 90% of the disease
can be removed and the tumor is of low or interme-
diate grade, if the tumor location is causing prob-
lems, or if removal will reduce hormonal symptoms
2.7 MEtaStatIC NEt (Alagusundaramoorthy and Gedaly, 2014).
Surgery does not eliminate the metastatic dis-
NETs are a heterogenous group of neoplasms that ease but offers the best achievable tumor control.
originate from foregut structures (lung and pan- A retrospective international study compared
creas), midgut (small intestine and part of colon), surgery with transarterial therapy. This was not
and hindgut (distal colon). NET can be hormon- a randomized study and therefore was subjected
ally functional or nonfunctional depending on the to profound patient selection biases (the surgical
presence of clinical symptoms of hormone/peptide group had fewer hormonally active tumors and the
secretion (insulin, glucagon, gastrin, vasoactive overall hepatic burden was greater in the transar-
intestinal peptide, serotonin). Abdominal carci- terial group). The study found that the median and
noid tumors usually originate in the bowel and 5-year survival of patients treated with surgery was
present with typical symptoms of flushing, diar- 123 months and 74% versus 34 months and 30%
rhea, wheezing, or endocardial and valvular heart for transarterial therapy, a highly significant dif-
disease. Pancreatic and bowel NET have a strong ference. Surgery showed the greatest survival ben-
tendency for development of liver metastases, efit in symptomatic patients with a larger tumor
46 Treatment options for patients with primary and secondary liver cancer

burden, but when the patients were asymptom- morbidity and 5-year survival rates ranging from
atic, there was no difference in long-term outcome 57%–80%. Partial or complete symptom relief was
between surgery and transarterial therapy (Mayo achieved in over 90% of cases (Mohan et al., 2015).
et al., 2011).
In another study of hepatic resection, the over-
all survival at 5 and 10 years was 63% and 40%, 2.7.3 SYSTEMIC THERAPY FOR NET
respectively, but most patients had disease pro- Systemic therapy for metastatic NET can be directed
gression during follow-up. The authors suggested to target the hormonal/peptide-producing nature of
that aggressive surgery is beneficial for well- the tumor or the tumor growth. Synthetic soma-
differentiated metastatic tumors. Overall sur- tostatin analogs (SSA) are administered to most
vival confirmed large differences between those patients because most NET overexpress somatosta-
with well-, moderately, and poorly differentiated tin receptors and frequently produce hormonal/pep-
tumors, with median overall survival of approxi- tide symptoms. Even in tumors that do not present
mately 120, 60, and 20 months, respectively. The with clinical hormonal effects, SSA frequently have
presence of extrahepatic disease was also a predic- an antitumor effect with slowing of disease progres-
tor of poor prognosis (Saxena et al., 2011). sion and even tumor volume reduction. Octreotide
Liver transplantation is an option for selected is the most widely prescribed SSA. The PROMID
patients with NET liver metastases. When strin- clinical trial of long-acting octreotide showed an
gent selection criteria are applied, the results can antitumor effect, with a median time to progression
be excellent, with 5-year survival close to 90% and of 14.3 months for the experimental group, com-
recurrence-free survival close to 80%, but most pared with 6 months for placebo (Rinke et al., 2009).
patients do not qualify (de Herder et al., 2010). For tumor growth control, various chemo-
A more realistic analysis of the European Liver therapeutic agents are used including strepto-
Transplant Registry reported 5-year overall and zocin, dacarbazine, 5-fluorouracil, doxorubicin,
disease-free survival rates of 52% and 30%, respec- etoposide, cisplatin, carboplatin, and taxanes.
tively (Le Treut et al., 2013). Targeted therapies are valuable, the most useful
of which include the tyrosine kinase inhibitors
2.7.2 TUMOR ABLATION FOR NET sunitinib and sorafenib, the VEGF monoclonal
antibody bevacizumab, and the mammalian target
Tumor ablation can be a stand-alone therapy, an of rapamycin (mTOR) inhibitor everolimus. The
adjunct at the time of surgical resection, for recur- development of β-emitting radionuclide-labeled
rence after previous ablation or in patients who somatostatin analogs 90Y-DOTATOC (DOTA0-
cannot have surgery because of comorbidities and D-Phe1-Tyr3-octreotide) and 177Lu-DOTATATE
other risk factors. Ablation is most valuable when (DOTA-(Tyr3)-octreotate) offers targeted radiation
metastases are few and <5 cm in diameter (ideally therapy with a high degree of tissue specificity.
<3 cm) and if disease is in a location where adjacent In general, sunitinib or everolimus are first-
tissues would not be compromised. Intraoperative line options for symptomatic pancreatic, bulky,
tumor ablation can address disease that is found or progressive NET. For symptomatic, bulky, or
beyond the surgical margin and has been found to progressive gastroenteric carcinoid tumors, the
widen the patient eligibility for surgery and provide SSA are first-line choices. Where available, the
additional symptom control (Taner et al., 2013). radionuclide-labeled SSA can also be used as a
Outcomes data for percutaneous ablation first-line therapy (Castellano et al., 2015).
of metastatic NET are limited by the absence of
randomized trials, the selection biases that are
inherent to the assignment of surgical versus 2.7.4 TRANSARTERIAL THERAPY
percutaneous approaches, and the mixed nature FOR NET
of reports that combine both intraoperative and
percutaneous ablation. A recent meta-analysis of Transarterial therapies are commonly applied
301 patients and 978 tumors confirmed the pro- to nonsurgical patients with liver-dominant dis-
cedure’s safety with a 0.7% mortality rate and 10% ease to control tumor growth and symptoms.
References 47

Virtually any embolic agent will be effective by Gemcitabine is the most commonly used agent,
inducing ischemia, delivering high-dose chemo- along with 5-fluorouracil and cisplatin. It is still
therapy, or delivering intra-arterial radioactive early to determine whether targeted therapies
beads. The data are insufficient to demonstrate impact the outcomes of ICC (Lafaro et al., 2015).
any advantage between inert particles, drug-elut- External-beam radiation can be used follow-
ing beads, or conventional lipiodol-based TACE ing surgery or as the primary therapy, but stud-
(Brown et al., 1999; Orgera et al., 2015). A study of ies are small and not controlled. As the primary
100 patients with metastatic NET showed median therapy, the median overall survival can be as high
overall survival of approximately two years (Pitt as 13.3 months (Ben-Josef et al., 2005). SBRT can
et al., 2008). be more effective, but fewer patients qualify for
The available trials do not show superiority of this approach. A study from Mayo Clinic showed
any technique, but doxorubicin DEB-TACE has an overall survival at 6 and 12 months of 83% and
been associated with increased risk for biliary 73%, but this was for a very small, highly selected
injury and complications (Bhagat et al., 2013). In a patient group (Barney et al., 2012). The use of
retrospective series, the median survival of meta- SBRT for ICC is becoming more widely accepted
static NET treated with Y-90 microspheres was 70 (Tanguturi et al., 2014).
months (Kennedy et al., 2008). Direct compari- Radioembolization with Y-90 microspheres has
son outcomes data between radioembolization shown effectiveness comparable to systemic che-
with other transarterial therapies do not exist, motherapy. A systematic review and pooled analy-
and the available data are heavily influenced by sis of 12 studies and 298 patients yielded a median
selection bias. survival of 15.5 months. An added benefit of radio-
embolization is the conversion of unresectable to
surgically resectable disease (Al-Adra et al., 2015).
ICC treated with TACE has been shown to have
2.8 INtraHEPatIC complete or partial response in 25% and is associ-
CHOLaNGIOCarCINOMa ated with improved survival (Hyder et al., 2013).
The median overall survival is 9.1 months after
ICC is the second most common primary liver TACE, but TACE done with a combination of gem-
malignancy after HCC and originates from bile citabine plus cisplatin may be more effective than
ducts of small size within the liver. Most ICCs are single-agent TACE with median survival up to 18.8
incurable, respond poorly to therapy, and recur months (Gusani et al., 2008).
early and often. ICC predominantly spreads in Tumor ablation is applicable only in a minority
the liver but commonly becomes extrahepatic of ICCs because of criteria for treatment overlap
and produces distant hematogenous metastases with surgery. Percutaneous ablation is limited only
(Lafaro et al., 2015). to those who are not deemed surgical candidates
Surgery can be curative only if the resection is if the size, lesion number, and location are appro-
complete. Even if preoperative imaging suggests priate. Small ICCs can have excellent response to
that a complete resection is possible, at the time of tumor ablation, with a median overall survival of
resection many tumors cannot be removed entirely, 33 months (Fu et al., 2012).
and the recurrence rate is high. About 30% of cases
are candidates for surgery, and the resections are REFERENCES
usually extensive and complex, often requiring
intervention on extrahepatic bile ducts and vascu- Abulkhir, A., Limongelli, P., Healey A.J. et al.
lar structures (Endo et al., 2008). The 5-year over- (2008). Preoperative portal vein embolization
all survival despite surgery is poor, ranging from for major liver resection: A meta-analysis. Ann
14%–40%, and the median disease-free survival is Surg 247:49–57.
approximately 1 year (Lafaro et al., 2015). Ahmed, M. (2014). Image-guided tumor ablation:
Unresectable patients receive systemic chemo- Standardization of terminology and report-
therapy and have a poor prognosis, with variable ing criteria—A 10-year update. Radiology
median survival that averages about 6 months. 273:241–260.
48 Treatment options for patients with primary and secondary liver cancer

Al-Adra, D.P., Gill, R.S., Axford, S.J. et al. (2015). Brown, D.B. (2011). Hepatic artery dissection in a
Treatment of unresectable intrahepatic patient on bevacizumab resulting in pseudoa-
cholangiocarcinoma with yttrium-90 radioem- neurysm formation. Semin Intervent Radiol
bolization: A systematic review and pooled 28:142–146.
analysis. Eur J Surg Oncol 41:120–127. Brown, K.T., Koh, B.Y., Brody, L.A. et al. (1999).
Alagusundaramoorthy, S.S., Gedaly, R. (2014). Particle embolization of hepatic neuroen-
Role of surgery and transplantation in the docrine metastases for control of pain and
treatment of hepatic metastases from neu- hormonal symptoms. J Vasc Interv Radiol
roendocrine tumor. World J Gastroenterol 10:397–403.
20:14348–14358. Bruix, J., Sherman, M. (2011). Management
Aliberti, C., Tilli, M., Benea, G., Fiorentini, of hepatocellular carcinoma: An update.
G. (2006). Trans-arterial chemoemboli- Hepatology 53:1020–1022.
zation (TACE) of liver metastases from Camma, C., Schepis, F., Orlando, A. et al. (2002).
colorectal cancer using irinotecan-eluting Transarterial chemoembolization for unresect-
beads: Preliminary results. Anticancer Res able hepatocellular carcinoma: Meta-analysis
26:3793–3795. of randomized controlled trials. Radiology
Amersi, F.F., McElrath-Garza, A., Ahmad, A. et al. 224:47–54.
(2006). Long-term survival after radiofrequency Castellano, D., Grande, E., Valle, J. et al.
ablation of complex unresectable liver tumors. (2015). Expert consensus for the management
Arch Surg 141:581–587; discussion 587–588. of advanced or metastatic pancreatic neu-
Aragon-Ching, J.B., Ning, Y.M., Dahut, W.L. roendocrine and carcinoid tumors. Cancer
(2008). Acute aortic dissection in a hyperten- Chemother Pharmacol 75:1099–1114.
sive patient with prostate cancer undergoing Cho, Y.K., Kim, J.K., Kim, W.T., Chung,
chemotherapy containing bevacizumab. Acta J.W. (2010). Hepatic resection ver-
Oncol 47:1600–1601. sus radiofrequency ablation for very
Barney, B.M., Olivier, K.R., Miller, R.C., Haddock, early stage hepatocellular carcinoma:
M.G. (2012). Clinical outcomes and toxicity A Markov model analysis. Hepatology
using stereotactic body radiotherapy (SBRT) 51:1284–1290.
for advanced cholangiocarcinoma. Radiat Davis, G.L., Alter, M.J., El-Serag, H. et al. (2010).
Oncol 7:67. Aging of hepatitis C virus (HCV)-infected
Ben-Josef, E., Normolle, D., Ensminger, persons in the United States: A multiple
W.D. et al. (2005). Phase II trial of high-dose cohort model of HCV prevalence and disease
conformal radiation therapy with concur- progression. Gastroenterology 138:513–521,
rent hepatic artery floxuridine for unresect- 521 e511–516.
able intrahepatic malignancies. J Clin Oncol de Herder, W.W., Mazzaferro, V., Tavecchio, L.
23:8739–8747. et al. (2010) Multidisciplinary approach for the
Benson AB, 3rd et al. (2014). Colon cancer, treatment of neuroendocrine tumors. Tumori
version 3.2014. J Natl Compr Canc Netw 96:833–846.
12:1028–1059. Eghtesad, B., Aucejo, F. (2014) Liver transplanta-
Bhagat, N., Reyes, D.K., Lin, M. et al. (2013). tion for malignancies. J Gastrointest Cancer
Phase II study of chemoembolization with 45:353–362.
drug-eluting beads in patients with hepatic Endo, I., Gonen, M., Yopp, A.C. et al. (2008).
neuroendocrine metastases: High incidence Intrahepatic cholangiocarcinoma: Rising
of biliary injury. Cardiovasc Intervent Radiol frequency, improved survival, and determi-
36:449–459. nants of outcome after resection. Ann Surg
Boutros, C., Somasundar, P., Garrean S. et al. 248:84–96.
(2010). Microwave coagulation therapy for Erinjeri, J.P., Clark, T.W. (2010). Cryoablation:
hepatic tumors: Review of the literature and Mechanism of action and devices. J Vasc
critical analysis. Surg Oncol 19:e22–e32. Interv Radiol 21:S187–S191.
References 49

Ferlay, J., Shin, H.R., Bray, F. et al. (2010). Guiu, B., Bize, P., Gunthern, D. et al. (2013). Portal
Estimates of worldwide burden of cancer vein embolization before right hepatectomy:
in 2008: GLOBOCAN 2008. Int J Cancer Improved results using n-butyl-cyanoacrylate
127:2893–2917. compared to microparticles plus coils.
Fernandez-Ros, N., Silva, N., Bilbao, J.I. et al. Cardiovasc Intervent Radiol 36:1306–1312.
(2014). Partial liver volume radioembolization Gusani, N.J., Balaa, F.K., Steel, J.L. et al. (2008).
induces hypertrophy in the spared hemiliver Treatment of unresectable cholangiocarci-
and no major signs of portal hypertension. noma with gemcitabine-based transcatheter
HPB (Oxford) 16:243–249. arterial chemoembolization (TACE): A single-
Fiorentini, G., Aliberti, C., Tilli, M. et al. (2012). institution experience. J Gastrointest Surg
Intra-arterial infusion of irinotecan-loaded 12:129–137.
drug-eluting beads (DEBIRI) versus intrave- Hong, K., Georgiades, C. (2010) Radiofrequency
nous therapy (FOLFIRI) for hepatic metasta- ablation: Mechanism of action and
ses from colorectal cancer: Final results of a devices. J Vasc Interv Radiol 21:S179–S186.
phase III study. Anticancer Res 32:1387–1395. Hyder, O., Marsh, J.W., Salem, R. et al. (2013).
Forner, A., Vilana, R., Ayuso, C. et al. (2008). Intra-arterial therapy for advanced intra-
Diagnosis of hepatic nodules 20 mm or hepatic cholangiocarcinoma: A multi-
smaller in cirrhosis: Prospective validation of institutional analysis. Ann Surg Oncol
the noninvasive diagnostic criteria for hepato- 20:3779–3786.
cellular carcinoma. Hepatology 47:97–104. Ito, K., Govindarajan, A., Ito, H., Fong, Y. (2010).
Fu, Y., Yang, W., Wu, W. et al. (2012). Surgical treatment of hepatic colorectal
Radiofrequency ablation in the management metastasis: Evolving role in the setting of
of unresectable intrahepatic cholangiocarci- improving systemic therapies and ablative
noma. J Vasc Interv Radiol 23:642–649. treatments in the 21st century. Cancer J
Garlipp, B., de Baere, T., Damm, R. et al. (2014). 16:103–110.
Left-liver hypertrophy after therapeutic Kabbinavar, F.F., Hambleton, J., Mass, R.D. et al.
right-liver radioembolization is substantial (2005). Combined analysis of efficacy: The
but less than after portal vein embolization. addition of bevacizumab to fluorouracil/leu-
Hepatology 59:1864–1873. covorin improves survival for patients with
Gehl, J. (2003). Electroporation: Theory and metastatic colorectal cancer. J Clin Oncol
methods, perspectives for drug delivery, 23:3706–3712.
gene therapy and research. Acta Physiol Kennedy, A.S., Dezarn, W.A., McNeillie, P. et al.
Scand 177:437–447. (2008). Radioembolization for unresectable
Gibbs, P., Gebski, V., Van Buskirk, M. et al. (2014). neuroendocrine hepatic metastases using
Selective internal radiation therapy (SIRT) with resin 90Y-microspheres: Early results in 148
yttrium-90 resin microspheres plus standard patients. Am J Clin Oncol 31:271–279.
systemic chemotherapy regimen of FOLFOX Kim, R.D., Kim, J.S., Watanabe, G. et al.
versus FOLFOX alone as first-line treatment (2008). Liver regeneration and the atrophy-
of non-resectable liver metastases from hypertrophy complex. Semin Intervent Radiol
colorectal cancer: The SIRFLOX study. BMC 25:92–103.
Cancer 14:897. Kim, W.R., Gores, G.J., Benson, J.T. et al. (2005).
Gordon, C.R., Rojavin, Y., Patel, M. et al. (2009). Mortality and hospital utilization for hepa-
A review on bevacizumab and surgical wound tocellular carcinoma in the United States.
healing: An important warning to all sur- Gastroenterology 129:486–493.
geons. Ann Plast Surg 62:707–709. Kishi, Y., Abdalla, E.K., Chun, Y.S. et al. (2009).
Groeschl, R.T., Pilgrim, C.H., Hanna, E.M. et al. Three hundred and one consecutive extended
(2014). Microwave ablation for hepatic malig- right hepatectomies: Evaluation of outcome
nancies: a multiinstitutional analysis. Ann Surg based on systematic liver volumetry. Ann Surg
259:1195–1200. 250:540–548.
50 Treatment options for patients with primary and secondary liver cancer

Komorizono, Y., Oketani, M., Sako, K. et al. unresectable hepatocellular carcinoma: A ran-
(2003). Risk factors for local recurrence of domised controlled trial. Lancet 359:1734–1739.
small hepatocellular carcinoma tumors after Llovet, J.M., Ricci, S., Mazzaferro, V. et al. (2008).
a single session, single application of per- Sorafenib in advanced hepatocellular carci-
cutaneous radiofrequency ablation. Cancer noma. N Engl J Med 359:378–390.
97:1253–1262. Lo, C.M., Ngan, H., Tso, W.K. et al. (2002).
Kunz, P.L. (2015). Carcinoid and neuroendocrine Randomized controlled trial of transarterial
tumors: Building on success. J Clin Oncol lipiodol chemoembolization for unresect-
33:1855–1863. able hepatocellular carcinoma. Hepatology
Lafaro, K.J., Cosgrove, D., Geschwind, J.F. et al. 35:1164–1171.
(2015). Multidisciplinary care of patients with Maluccio, M.A., Covey, A.M., Porat, L.B. et al.
intrahepatic cholangiocarcinoma: Updates (2008). Transcatheter arterial emboliza-
in management. Gastroenterol Res Pract tion with only particles for the treatment of
2015:860861. unresectable hepatocellular carcinoma. J Vasc
Lammer, J., Malagari, K., Vogl, T. et al. (2010). Interv Radiol 19:862–869.
Prospective randomized study of doxorubi- Mantia-Smaldone, G.M., Bagley, L.J., Kasner, S.E.,
cin-eluting-bead embolization in the treat- Chu, C.S. (2013). Vertebral artery dissection
ment of hepatocellular carcinoma: Results of and cerebral infarction in a patient with recur-
the PRECISION V study. Cardiovasc Intervent rent ovarian cancer receiving bevacizumab.
Radiol 33:41–52. Gynecol Oncol Case Rep 5:37–39.
Le Treut, Y.P., Grégoire, E., Klempnauer, J. et al. Marrero, J.A., Hussain, H.K., Nghiem, H.V. et al.
(2013) Liver transplantation for neuroendo- (2005). Improving the prediction of hepatocel-
crine tumors in Europe-results and trends lular carcinoma in cirrhotic patients with an
in patient selection: A 213-case European arterially-enhancing liver mass. Liver Transpl
liver transplant registry study. Ann Surg 11:281–289.
257:807–815. Mayo, S.C. et al. (2011) Surgery versus intra-
Lencioni, R. (2010). Loco-regional treatment arterial therapy for neuroendocrine liver
of hepatocellular carcinoma. Hepatology metastasis: A multicenter international analy-
52:762–773. sis. Ann Surg Oncol 18:3657–3665.
Lencioni, R., Crocetti, L., Narayanan, G. (2015). Minocha, J., Salem, R., Lewandowski, R.J. (2014).
Irreversible electroporation in the treatment Transarterial chemoembolization and yit-
of Hepatocellular carcinoma. Tech Vasc Interv trium-90 for liver cancer and other lesions.
Radiol 18:135–139. Clin Liver Dis 18:877–890.
Lewandowski, R.J., Davenport, M.S. (2015). Mohan, H., Nicholson, P., Winter, D.C. et al.
Imaging and image-guided intervention are (2015). Radiofrequency ablation for neuroen-
irrevocably linked. Radiol Clin North Am 53:xi. docrine liver metastases: A systematic review.
Liapi, E., Geschwind, J.F. (2010). Intra-arterial J Vasc Interv Radiol 26:935–942.
therapies for hepatocellular carcinoma: Where Molvar, C., Lewandowski, R.J. (2015). Intra-
do we stand? Ann Surg Oncol 17:1234–1246. arterial therapies for liver masses: Data dis-
Livraghi, T., Solbiati, L., Meloni, M.F. et al. tilled. Radiol Clin North Am 53:973–984.
(2003). Treatment of focal liver tumors with Moreno-Luna, L.E., Yang, J.D., Sanchez,
percutaneous radio-frequency ablation: W. et al. (2013). Efficacy and safety of transarte-
Complications encountered in a multicenter rial radioembolization versus chemoemboliza-
study. Radiology 226:441–451. tion in patients with hepatocellular carcinoma.
Llovet, J.M., Bru, C., Bruix, J. (1999). Prognosis of Cardiovasc Intervent Radiol 36:714–723.
hepatocellular carcinoma: The BCLC staging Muratore, A., Zorzi, D., Bouzari, H. et al. (2007).
classification. Semin Liver Dis 19:329–338. Asymptomatic colorectal cancer with
Llovet, J.M., Real, M.I., Montana, X. et al. (2002). unresectable liver metastases: Immediate
Arterial embolisation or chemoembolisation colorectal resection or up-front systemic che-
versus symptomatic treatment in patients with motherapy? Ann Surg Oncol 14:766–770.
References 51

Orgera G, Krokidis M, Cappucci M. et al. (2015). advanced neuroendocrine neoplasms (NENs)


Current status of interventional radiology in supports an aggressive surgical approach.
the management of gastro-entero-pancre- Surgery 149:209–220.
atic neuroendocrine tumours (GEP-NETs). Scappaticci, F.A., Fehrenbacher, L., Cartwright,
Cardiovasc Intervent Radiol 38:13–24. T. et al. (2005). Surgical wound healing
Peng, Z.W., Chen, M.S., Liang, H.H. et al. (2010). complications in metastatic colorectal cancer
A case-control study comparing percutaneous patients treated with bevacizumab. J Surg
radiofrequency ablation alone or combined Oncol 91:173–180.
with transcatheter arterial chemoemboliza- Scheffer, H.J., Vroomen, L.G., Nielsen, K. et al.
tion for hepatocellular carcinoma. Eur J Surg (2015). Colorectal liver metastatic disease:
Oncol 36:257–263. Efficacy of irreversible electroporation—A
Pitt, S.C., Knuth, J., Keily, J.M. et al. (2008). single-arm phase II clinical trial (COLDFIRE-2
Hepatic neuroendocrine metastases: Chemo- trial). BMC Cancer 15:772.
or bland embolization? J Gastrointest Surg Shibata, T., Niinobu, T., Ogata, N.,
12:1951–1960. Takami, M. (2000). Microwave coagulation
Raman, S.S., Aziz, D., Chang, X. et al. (2004). therapy for multiple hepatic metastases from
Minimizing central bile duct injury dur- colorectal carcinoma. Cancer 89:276–284.
ing radiofrequency ablation: Use of intra- Shindoh, J., Tzeng, C.W., Aloia, T.A. et al. (2013b).
ductal chilled saline perfusion–Initial Portal vein embolization improves rate of
observations from a study in pigs. Radiology resection of extensive colorectal liver metas-
232:154–159. tases without worsening survival. Br J Surg
Ratti, F., Soldati, C., Catena, M. et al. (2010). Role 100:1777–1783.
of portal vein embolization in liver surgery: Shindoh, J., Vauthey, J.N., Zimmitti, G. et al.
Single centre experience in sixty-two patients. (2013a). Analysis of the efficacy of portal
Updates Surg 62:153–159. vein embolization for patients with extensive
Raut, C.P., Izzo, F., Marra, P. et al. (2005). liver malignancy and very low future liver
Significant long-term survival after radiofre- remnant volume, including a comparison with
quency ablation of unresectable hepatocel- the associating liver partition with portal vein
lular carcinoma in patients with cirrhosis. ligation for staged hepatectomy approach. J
Ann Surg Oncol 12:616–628. Am Coll Surg 217:126–133; discussion 133–124.
Reig, M., Darnell, A., Forner, A. et al. (2014). Siegel, R., Naishadham, D., Jemal, A. (2013).
Systemic therapy for hepatocellular carcinoma: Cancer statistics, 2013. CA Cancer J Clin
The issue of treatment stage migration and 63:11–30.
registration of progression using the BCLC- Silk, M., Tahour, D., Srimathveeravalli,
refined RECIST. Semin Liver Dis 34:444–455. G. et al. (2014). The state of irreversible elec-
Riaz, A., Lewandowski, R.J., Kulik, L.M. et al. troporation in interventional oncology. Semin
(2009). Complications following radioem- Intervent Radiol 31:111–117.
bolization with yttrium-90 microspheres: A Simon, C.J., Dupuy, D.E., Mayo-Smith, W.W. (2005)
comprehensive literature review. J Vasc Interv Microwave ablation: Principles and applica-
Radiol 20:1121–1130; quiz 1131. tions. Radiographics 25(Suppl 1):S69–S83.
Rinke, A., Muller, H.H., Schade-Brittinger, C. et Simoneau, E., Aljiffry, M., Salman, A. et al.
al. (2009). Placebo-controlled, double-blind, (2012). Portal vein embolization stimulates
prospective, randomized study on the effect of tumour growth in patients with colorec-
octreotide LAR in the control of tumor growth tal cancer liver metastases. HPB (Oxford)
in patients with metastatic neuroendocrine 14:461–468.
midgut tumors: A report from the PROMID Solbiati, L., Ierace, T., Goldberg, S.N. et al. (1997).
Study Group. J Clin Oncol 27:4656–4663. Percutaneous US-guided radio-frequency
Saxena, A., Chua, T.C., Sarkar, A. et al. (2011). tissue ablation of liver metastases: Treatment
Progression and survival results after radi- and follow-up in 16 patients. Radiology
cal hepatic metastasectomy of indolent 202:195–203.
52 Treatment options for patients with primary and secondary liver cancer

Takaki, H., Yamakado, K., Uraki, J. et al. (2009). colorectal liver metastases. Eur J Cancer
Radiofrequency ablation combined with che- 42:2212–2221.
moembolization for the treatment of hepato- Vogl, T.J., Farshid, P., Naguib, N.N. et al. (2015).
cellular carcinomas larger than 5 cm. J Vasc Ablation therapy of hepatocellular carcinoma:
Interv Radiol 20:217–224. a comparative study between radiofrequency
Taner, T., Atwell, T.D., Zhang, L. et al. (2013). and microwave ablation. Abdom Imaging
Adjunctive radiofrequency ablation of meta- 40:1829–1837.
static neuroendocrine cancer to the liver Wells, S.A., Hinshaw, J.L., Lubner, M.G. et al.
complements surgical resection. HPB (Oxford) (2015). Liver ablation: Best practice. Radiol
15:190–195. Clin North Am 53:933–971.
Tanguturi, S.K., Wo, J.Y., Zhu, A.X. et al. (2014) Yao, J.C., Hassan, M., Phan, A. et al. (2008).
Radiation therapy for liver tumors: Ready One hundred years after “carcinoid”:
for inclusion in guidelines? Oncologist Epidemiology of and prognostic fac-
19:868–879. tors for neuroendocrine tumors in 35,825
Teo, J.Y., Goh, B.K. (2015). Contra-lateral cases in the United States. J Clin Oncol
liver lobe hypertrophy after unilobar Y90 26:3063–3072.
radioembolization: An alternative to portal Yoo, P.S., Lopez-Soler, R.I., Longo, W.E., Cha,
vein embolization? World J Gastroenterol C.H. (2006). Liver resection for metastatic
21:3170–3173. colorectal cancer in the age of neoadjuvant
Teratani, T., Yoshida, H., Shiina, S. et al. (2006). chemotherapy and bevacizumab. Clin
Radiofrequency ablation for hepatocellular Colorectal Cancer 6:202–207.
carcinoma in so-called high-risk locations. Ziemlewicz, T.J., Hinshaw, J.L., Lubner, M.G. et al.
Hepatology 43:1101–1108. (2015). Percutaneous microwave ablation of
Van Cutsem, E., Nordlinger, B., Adam, R. hepatocellular carcinoma with a gas-cooled
et al. (2006). Towards a pan-European system: initial clinical results with 107 tumors.
consensus on the treatment of patients with J Vasc Interv Radiol 26:62–68.
3
Treatment planning part I: Vascular
considerations associated with safety
and efficacy in radioembolization

RAY BRADFORD AND J. MARK MCKINNEY

3.1 Introduction 53 3.7 Techniques to minimize


3.2 Arterial access 54 hepatopulmonary shunting 59
3.3 Hepatic arterial anatomy 54 3.8 Complications 60
3.4 Complicated hepatic arterial access 56 3.9 Angiogenesis 61
3.5 Parasitized arterial perfusion 57 3.10 Conclusion 61
3.6 Techniques to prevent nontarget References 61
radioembolization 58

The arteries perfusing the liver also supply


3.1 INtrODUCtION many other important visceral structures includ-
ing the stomach, duodenum, esophagus, pancreas,
Yttrium-90 (90Y) radioembolization is delivered gallbladder, and abdominal wall. Consideration
via arterial supply to the liver. Hepatic neo- must be given to protection against nontarget
plasms receive greater than 80% of their perfu- embolization of these extrahepatic visceral struc-
sion from the hepatic arteries, while the normal tures. Nontarget embolization is avoided during
hepatic parenchyma receives the majority of its radioembolization with several techniques, such as
blood supply from the portal vein. This differen- adjusting the catheter tip location, employing anti-
tial blood supply allows for the arterial delivery reflux catheters, and/or using protective embolic
of relatively large radiation doses to the tumor redistribution of arterial flow.
with relative sparing of normal liver parenchyma In addition to the classic and variant arterial
(Welsh et al., 2006). 90Y radioembolization for vascular supply to the liver, the interventionalist
primary and/or secondary neoplasms of the liver must take into account extrahepatic arterial para-
necessitates a thorough understanding of hepatic sitization, which occurs with large or peripheral
arterial anatomy to ensure safety and efficacy. The hypervascular neoplasms. Extrahepatic arteries are
interventionalist must be aware that variations in parasitized from multiple vascular distributions in
the arterial supply to the liver are common and proximity to the liver parenchyma. Effective 90Y
that these anatomic variations affect both hepatic radioembolization may require catheter-directed
lobes. therapy to these nonhepatic arterial distributions.
53
54 Treatment planning part I

Arterial delivery of 90Y radioembolization requi-


res both training and experience because anatomic
treatment decisions are often complex and can be
complicated by anatomic variations, extent of dis-
ease, and prior treatments. This chapter is not meant
to cover every detail of all vascular aspects related to
radioembolization, but rather it is meant to provide LHA

an overview that can be appreciated and understood


by all members of the radioembolization team. RHA PHA CHA
GDA

3.2 artErIaL aCCESS


Figure 3.1 Celiac arteriogram demonstrates
Interventionalists have the option to approach the classic hepatic arterial anatomy. The common
hepatic arterial supply via a femoral or radial artery. hepatic artery (CHA) continues as the proper
hepatic artery (PHA) beyond the takeoff of the
Femoral artery access is historically the most com-
gastroduodenal artery (GDA). The PHA splits into
mon access point for diagnostic and therapeutic
the right hepatic artery (RHA) and left hepatic
arterial interventions. However, with the advances artery (LHA).
of lower profile catheters and the desire to improve
patient satisfaction and comfort with early ambu- RHA (6%) from the superior mesenteric artery
lation, radial artery access has become the favored (Covey et al., 2002; Lee et al., 2012). Variations
access point for many interventionalists. Prior to of the left hepatic artery (LHA) supply include
radial artery puncture, patency of the ulnar artery replacement of the LHA (5%) and accessory LHA
and collateral supply through the palmar arch is (15%) from the left gastric artery (LGA) (Covey et
confirmed utilizing a Barbeau test (Barbeau et al., al., 2002; Lee et al., 2012) (Figure 3.2). In addition,
2004). The risk of radial artery spasm and throm- a middle hepatic artery may present either as an
bosis is mitigated via intra-arterial infusion of hep- accessory branch of the RHA or a true trifurcation
arin, verapamil, and nitroglycerin (Bishay et al., from the PHA (Kerlan and LaBerge, 2006).
2014). A Glidesheath is also utilized to minimize Extrahepatic arteries are important for the
trauma to the radial artery. Approaching the celiac interventionalist utilizing 90Y radioemboliza-
artery from a radial artery approach may have ana- tion as locoregional therapy. Extrahepatic arter-
tomic advantages during celiac and hepatic arte- ies are a potential nontarget pathway for 90Y to be
rial catheter placement. Potential disadvantages diverted from the targeted hepatic parenchyma to
to radial artery access include the need to utilize nontargeted adjacent viscera or musculoskeletal
longer catheter delivery systems and ergonomic structures. Specific extrahepatic arteries at risk for
challenges for the operator. nontarget radioembolization typically include the
right gastric, gastroduodenal, pancreaticoduode-
nal, cystic, esophageal, and falciform arteries. Pre-
90Y radioembolization planning arteriography is
3.3 HEPatIC artErIaL
utilized to map and discover potential perihepatic
aNatOMY arterial pathways that place the patient at risk for
nontarget embolization during 90Y therapy.
Classic hepatic arterial supply arises from the celiac The right gastric artery (RGA) frequently arises
artery with bifurcation of the proper hepatic artery from LHA, PHA, gastroduodenal artery (GDA),
(PHA) into the right and left intrahepatic arterial or common hepatic artery (CHA) (VanDamme
branches (Figure 3.1). This classic pattern is esti- and Bonte, 1990; Liu et al., 2005). Because both
mated to be present in greater than 60% of patients RGA and LGA perfuse the lesser curvature of
(Covey et al., 2002; Lee et al., 2012). Variations of the stomach, it is important to identify the RGA
the right hepatic arterial supply include replaced origin. RGA is characteristically small in cali-
right hepatic artery (RHA) (12%) and accessory ber and may have a sharply angulated origin
3.3 Hepatic arterial anatomy 55

LGHA
LGHA

(a) (b)

Figure 3.2 Variant hepatic arterial anatomy. (a) Replaced right hepatic artery from the SMA. (b) Left
gastrohepatic artery (LGHA) with branches to gastric fundus (arrowheads) and left hepatic lobe (arrow).

LGA
LHA

RGA

(a) (b)

Figure 3.3 Left and right gastric arteries. (a) Left gastric arteriogram demonstrates cross filling from
the left gastric artery (LGA) to the right gastric artery (RGA) which arises from the left hepatic artery
(LHA). (b) Direct selective right gastric arteriogram in a different patient.

that makes selective catheterization challenging. of nontarget embolization to the gastroduode-


When the RGA origin cannot be identified, it is nal artery must be evaluated due to its continuity
frequently evaluated via a left gastric arteriogram with the PHA and the subsequent bifurcation of
(Figure 3.3). the PHA into the RHA and LHAs. Depending on
The gastroduodenal artery is a relatively large treatment intent and catheter tip location for 90Y
artery arising from the CHA and supplies the radioembolization, the gastroduodenal artery may
pancreas, duodenum, and greater curvature of not be at significant risk.
the stomach via the pancreaticoduodenal arcade The cystic artery perfuses the gallbladder and
and gastroepiploic arteries (Figure 3.1). The risk typically originates from the proximal RHA.
56 Treatment planning part I

Clinical symptoms from radioembolic cholecysti- pathways that can complicate radioembolization
tis can occur but are usually self-limiting. include the esophageal and gastric branches aris-
Specific attention to the intrahepatic arterial ing from the LHA (Figure 3.5) and the duodenal
distribution is important for evaluating several branches arising from the central hepatic arter-
extrahepatic arterial perfusion pathways. One of ies. With careful analysis, these extrahepatic
these intrahepatic-to-extrahepatic arterial path- arterial pathways can be identified and strategies
ways is the falciform artery, which arises from the can be developed to protect against nontarget 90Y
left or middle hepatic arteries and perfuses the radioembolization.
anterior abdominal wall (Figure 3.4) (Baba et al., Some posttreatment examples of 90Y nontar-
2000; Liu et al., 2005). Other extrahepatic artery get embolization through extrahepatic arteries
are shown in Chapter 13, associated with clinical
sequelae discussed in Chapter 14.

3.4 COMPLICatED HEPatIC


artErIaL aCCESS

Diffuse or focal vascular disease may be a com-


plicating factor regardless of whether the inter-
ventionalist utilizes a femoral or radial arterial
approach. Severe peripheral vascular disease with
atherosclerotic stenosis of the aorta or iliac arter-
ies may require a contralateral femoral artery
approach or radial artery approach. Prior to choice
of arterial access, the interventionalist should also
be aware of the patient’s prior surgical history,
which may include aortic, iliac, femoral, or upper
extremity surgical grafts.
Figure 3.4 Falciform artery (arrow) arises from Celiac artery stenosis from median arcuate lig-
the left hepatic artery. ament syndrome (Figure 3.6) or atherosclerosis

(a) (b)

Figure 3.5 Esophageal artery. (a) An esophageal artery (arrowheads) arises from the left hepatic
arteries (arrows). (b) Coil embolization (arrow) of the esophageal branches to prevent nontarget
radioembolization.
3.5 Parasitized arterial perfusion 57

GDA

SMA

PDA

Figure 3.7 Occluded celiac artery (arrowhead).


Retrograde microcatheter access (curved arrows)
Figure 3.6 Median arcuate ligament narrowing to the hepatic arteries is possible via the superior
(arrow) the celiac artery. mesenteric artery (SMA), pancreaticoduodenal
arcade (PDA), and gastroduodenal artery (GDA).

may be encountered as a complicating access


issue. Stenosis from median arcuate ligament
3.5 ParaSItIZED artErIaL
syndrome is often incomplete and allows coax-
ial microcatheter advancement from an access
PErFUSION
catheter seated in the narrowed celiac artery.
Ultimate relief of median arcuate ligament syn- Consideration must be given to extrahepatic arte-
drome is surgical release (Columbo et al., 2015). rial pathways that may be recruited and para-
In the case of atherosclerotic celiac artery steno- sitized for perfusion of intrahepatic neoplasms.
sis or occlusion, celiac artery access for radioem- Characteristics that increase the likelihood
bolization can be achieved via celiac artery stent of parasitization include peripheral and large
placement. tumors and prior hepatic arterial embolization
In the cases where celiac occlusion is complete (Abdelmaksoud et al., 2011).
and celiac catheter access cannot be achieved, Awareness and identification of parasitized
enlarged pancreaticoduodenal arterial collater- extrahepatic arteries is necessary to completely
als from the superior mesenteric artery provide a treat targeted tumor beds. Tumors that receive
retrograde approach to hepatic artery 90Y radio- supplemental arterial blood supply from parasitized
embolization (Figure 3.7). To achieve hepatic extrahepatic arteries are particularly at risk of being
artery access via the pancreaticoduodenal col- undertreated (Abdelmaksoud et al., 2011). Bland,
laterals, an access catheter is seated in the supe- conventional chemoembolization, or drug-eluting
rior mesenteric artery and coaxial microcatheter bead chemoembolization of parasitized extrahe-
techniques are utilized to advance access serially patic arteries supplying peripherally located hepatic
through the superior mesenteric artery, inferior tumors provides therapeutic intent and reestablish-
pancreaticoduodenal trunk, pancreaticoduo- ment of primary hepatic arterial perfusion through
denal arcade, gastroduodenal artery, and into intrahepatic arteries. Extrahepatic arteries most
the proper and intrahepatic arterial branches. commonly recruited for tumor perfusion include
Supraselective access into the hepatic arteries the right inferior phrenic, internal mammary, inter-
may be limited when taking a circuitous retro- costal, right adrenal, right renal, and greater omen-
grade approach. tal arteries (Figure 3.8) (Abdelmaksoud et al., 2011).
58 Treatment planning part I

(a) (b)

(c) (d)

Figure 3.8 Parasitized extrahepatic arteries perfusing intrahepatic tumor (arrowheads). (a) Right infe-
rior phrenic artery. (b) Right internal mammary artery. (c) Intercostal artery. (d) Right adrenal and renal
arteries.

embolized at its origin to prevent nontarget embo-


3.6 tECHNIQUES tO lization. Gastroduodenal artery coil embolization
PrEVENt NONtarGEt is performed excluding the GDA as a potential
raDIOEMBOLIZatION pathway for nontarget embolization. However,
additional experience with 90Y radioembolization
has shown that gastroduodenal coil emboliza-
Coil embolotherapy is a well-developed technique
tion is frequently unnecessary and may actually
utilized by interventionalists to occlude and redi- increase the risk of intrahepatic recruitment of
rect arterial perfusion. Historically, coil embolo- duodenal and pancreatic arterial collateral path-
therapy is most frequently utilized in the GDA and ways (Hamoui et al., 2013a, 2013b). Avoidance of
RGA during planning arteriography to prevent gastroduodenal coil embolization has been dem-
nontarget embolization. onstrated to decrease procedure time, contrast
In the early implementation of radioemboliza- volume, and radiation exposure to the patient
tion, the gastroduodenal artery was routinely coil (Fischman et al., 2014).
3.7 Techniques to minimize hepatopulmonary shunting 59

The RGA is at a particular risk for nontarget phase because the need for coil embolotherapy is
radioembolization due to its proximity to typical eliminated or reduced (Fischman et al., 2014).
radioembolization catheter tip locations. The RGA
originates from either the LHA, PHA, GDA, or
CHA (Covey et al., 2002; Lee et al., 2012). The RGA
is often very small in caliber and has a sharply
3.7 tECHNIQUES tO MINIMIZE
angulated origin. When technically possible, the HEPatOPULMONarY
RGA is directly accessed via a coaxial microcath- SHUNtING
eter and its origin is coil embolized. When the
RGA cannot be identified or accessed directly, Hepatopulmonary shunting may lead to nontarget
it may be successfully approached via retrograde pulmonary embolization and must be recognized
access from the LGA. An access catheter is seated during planning and treatment with 90Y radio-
in the LGA origin, and a coaxial microcatheter is embolization. Hepatopulmonary shunt fraction
advanced along the communicating artery from is usually evaluated with a test dose of technium-
the left gastric to the RGA origin where coils are 99m microaggregated albumin (99mTc-MAA) dur-
carefully deposited. If 90Y catheter tip delivery does ing the arterial planning phase of 90Y treatment, as
not pose a risk to the RGA, then coil embolization described in Chapter 4. Arteriovenous shunting
is not necessary (Hamoui et al., 2013a, 2013b). into the hepatic veins and portal veins is common
Coil embolization is also utilized to protect other with liver tumors (Figure 3.9) (Sugano et al., 1994;
extrahepatic arterial beds such as the falciform Chan et al., 2010). 90Y microspheres can travel to
artery. If a falciform artery is identified (Figure 3.4), the pulmonary arterial bed via the hepatic and
coil embolization of the falciform artery is per- portal veins. Excessive hepatopulmonary shunt-
formed when technically possible because 90Y radio- ing with 90Y microspheres may in rare cases lead
embolization to the falciform artery can result in a to radiation pneumonitis. Radiation pneumoni-
highly localized midabdominal burning sensation tis manifests clinically with nonspecific symp-
for a period of days or weeks (Liu et al., 2005). When toms of fever, nonproductive cough, and dyspnea.
the falciform artery cannot be accessed, studies
have shown that ice packs to the anterior abdominal
wall provide vasoconstriction that reduces nontar-
get embolization to the terminal falciform arterial
branches (Wang et al., 2013).
Coil embolization is also applied when esopha-
geal or gastric branches are identified as origi-
nating within the intrahepatic arterial supply.
Branches to the esophagus and stomach may origi-
nate from the LHA and are embolized to prevent
nontarget embolization (Figure 3.5).
Antireflux catheters have been devised to mini-
mize nontarget embolization during radioembo-
lization (Figure 15.2). Catheters are designed to
deliver 90Y microspheres in target hepatic arter-
ies ranging from 2 to 6 mm in diameter. Studies
have demonstrated increased tumor uptake and
decreased nontarget embolization in multiple
tumor types (Pasciak et al., 2015). A prospective
randomized study of protective embolic coiling Figure 3.9 Arteriovenous shunting from hyper-
versus antireflux catheter delivery demonstrated vascular hepatocellular carcinoma leads to
reduced fluoroscopy time, procedure time, and early opacification of the draining hepatic vein
contrast dose during the planning arteriogram (arrowheads).
60 Treatment planning part I

Radiation pneumonitis is radiographically sug- Ward et al. (2015) now recommend that if the
gested as peribronchial cuffing on chest imaging expected lung dose is <30 Gy, no shunt mitigation
(Graves et al., 2010) and as a restrictive pattern on is required. For expected lung dose >30 Gy, cathe-
pulmonary function testing. Treatment is inhaled ter-based techniques can be utilized without delay
and/or systemic corticosteroids (Leung et al., to minimize nontarget radioembolization to the
1995). Ultimately, radiation pneumonitis can lead pulmonary arterial bed (Ward et al., 2015).
to debilitating chronic disease.
Because of the potential risks of an elevated pul-
monary shunt fraction leading to radiation pneu-
monitis, manufacturers have released guidelines 3.8 COMPLICatIONS
for 90Y microspheres. The resin 90Y microsphere
training manual guidelines by Sirtex Medical Complications from 90Y radioembolization have
(North Sydney, Australia) recommend a lung been reported in multiple studies. Early complica-
radiation dose limit of 25Gy per treatment ses- tions include fatigue, pain, nausea, emesis, and low-
sion, not to exceed a 50Gy cumulative dose. For grade fever. This constellation of early symptoms is
glass 90Y microspheres, the package insert by BTG typically called postembolization syndrome (Riaz
International (West Conshohocken, Pennsylvania) et al., 2009). Postembolization syndrome is usually
recommends an upper limit of 16.5 mCi deliv- self-limited and gradually resolves over the first
ered to the lungs. Ho et al. (1997) also recommend 1–2 weeks of treatment.
restricting the lung radiation absorbed dose to <30 Late complications of 90Y radioembolization
Gy. Dose reductions for an elevated hepatopulmo- include gastrointestinal ulceration, cholecystitis,
nary shunt fraction have been shown to result in pancreatitis, biliary injury, and radiation-induced
reduced efficacy of 90Y radioembolization therapy liver disease (Hamoui and Ryu, 2011), as well as
(Garin et al., 2015; Lam et al., 2015). However, pneumonitis. Gastrointestinal ulceration typi-
studies have shown that the risk from an elevated cally presents weeks after radioembolization as
hepatopulmonary shunt fraction is very low. In refractory abdominal pain, nausea, and vomiting.
one series, no patients were found to have radia- Gastrointestinal symptoms may be treated with
tion pneumonitis with a cumulative lung dose >30 proton pump inhibitors and sucralfate. Endoscopy
Gy (Salem et al., 2008). Additional considerations may be utilized to confirm the diagnosis of ulcer-
associated with hepatopulmonary shunt will be ation. Biopsy of the ulcers will often show micro-
discussed in Chapter 4. spheres in the biopsy specimen.
Several non–dose-reducing techniques have Radiation-induced liver disease typically occurs
been utilized to deal with high hepatopulmonary 4–8 weeks after radioembolization with elevation
shunt fraction. The use of systemic sorafenib treat- of alkaline phosphatase and bilirubin. Radiation-
ment has been shown to reduce hepatopulmo- induced liver disease is a clinical diagnosis asso-
nary shunt fraction by 62%–87% (Theysohn et ciated with ascites and jaundice (Sangro et al.,
al., 2012). Transarterial chemoembolization has 2008; Hamoui and Ryu, 2011). Multiple prior che-
resulted in reduction of hepatopulmonary shunt motherapy regimens are a risk factor for radia-
fraction by 25%–57% (Rose and Hoh, 2009; Gaba tion-induced liver disease. Dosimetric thresholds
and Vanmiddlesworth, 2012). The use of sorafenib related to radiation-induced liver disease are dis-
or transarterial chemoembolization may delay 90Y cussed in Chapter 5.
radioembolization; therefore, catheter-based tech- Biliary sequelae following 90Y radioemboliza-
niques to reduce shunting have been developed tion are usually clinically inconsequential. As with
for use during 90Y radioembolization rather than other liver-directed therapies, biliary complica-
reducing the treatment dose. Catheter-based tech- tions are seen more commonly with secondary
niques include temporary balloon occlusion of the neoplasms than with hepatocellular carcinoma.
hepatic veins or portal veins (Bester and Salem, Potential biliary complications included stric-
2007; Murata et al., 2009), embolization of varices, ture formation, obstruction, biloma, cholecysti-
or bland embolization of the hepatic tumor imme- tis, hepatic abscess, and serum bilirubin toxicity.
diately before or following 90Y radioembolization Patients are often asymptomatic, even with imag-
(Ward et al., 2015). ing evidence of biliary complications. Treatment
References 61

for biliary sequelae is based on clinical presenta- planning for variations of normal hepatic arterial
tion and may include antibiotics, percutaneous anatomy, considerable thought must be given to
drainage of fluid collections, biliary decompres- each specific patient’s situation. Unexpected com-
sion, and cholecystectomy (Atassi et al., 2008). plicating factors such as stenotic or occluded celiac
Additional discussion of the late complications of access, extrahepatic arterial communications, par-
radioembolization, including identification using asitized arterial perfusion, and hepatopulmonary
advanced imaging techniques, can be found in shunting are frequently encountered. Techniques
Chapters 13 and 14. such as coil embolization, antireflux catheters, and
dose modifications allow for safe and efficacious
delivery of yttrium-90 to primary and secondary
hepatic neoplasms.
3.9 aNGIOGENESIS
REFERENCES
Tumor growth and spread is known to be driven by
a complex interplay of proangiogenic and antian- Abdelmaksoud, M.H., Louie, J.D., Kothary, N. et al.
giogenic cytokines (Bergers and Benjamin, 2003). (2011). Embolization of parasitized extrahepatic
Since some patients experience early tumor recur- arteries to reestablish intrahepatic arterial sup-
rence following 90Y radioembolization, it is impor- ply to tumors before yttrium-90 radioemboliza-
tant to consider the role that cytokines may play. tion. J Vasc Interv Radiol 22:1355–1362.
Vascular endothelial growth factor (VEGF) levels Atassi, B., Bangash, A.K., Lewandowski, R.J. et al.
are known to be associated with suboptimal out- (2008). Biliary sequelae following radioembo-
comes in primary and secondary liver neoplasm. lization with yttrium-90 microspheres. J Vasc
In addition, VEGF is associated with hepatocellu- Interv Radiol 19:691–697.
lar carcinoma disease stage, presence of metastasis, Baba, Y., Miyazono, N., Ueno, K. et al. (2000).
vascular invasion, treatment response, and overall Hepatic falciform artery. Angiographic find-
survival (Xiong et al., 2004; Sergio et al., 2008). ings in 25 patients. Acta Radiol 41:329–333.
Carpizo et al. (2014) found that VEGF, angio- Barbeau, G.R., Arsenault, F., Dugas, L. et al.
poietin-2 (Ang-2), platelet-derived growth factor (2004). Evaluation of the ulnopalmar arterial
subunit BB (PDGF-BB), and other nonclassic cyto- arches with pulse oximetry and plethysmogra-
kines were temporally associated with 90Y radio- phy: Comparison with the Allen’s test in 1010
embolization. They observed spikes in the cytokine patients. Am Heart J 147:489–493.
baseline values as sampled following first- and Bergers, G., Benjamin, L.E. (2003). Tumorigenesis
second-stage 90Y radioembolization treatment epi- and the angiogenic switch. Nat Rev Cancer
sodes. This evidence suggests that 90Y radioembo- 3:401–410.
lization has the potential to upregulate angiogenic Bester, L., Salem, R. (2007). Reduction of arterio-
cytokines. When overall survival (OS) is evaluated hepatovenous shunting by temporary balloon
in association with cytokine release, there is corre- occlusion in patients undergoing radioemboli-
lation between shortened OS and temporal spikes zation. J Vasc Interv Radiol 18:1310–1314.
in VEGF, Ang-2, and PDGF-BB. These cytokines Bishay, V., Patel, R.S., Kim, E. et al. (2014).
appear to affect OS by promoting angiogenesis. It Transradial approach for hepatic radioembo-
is plausible that some patients might benefit from lization: Initial result and technique. J Vasc
antiangiogenic therapy administered before 90Y Interv Radiol 25:S88.
radioembolization (Carpizo et al., 2014). Carpizo, D.R., Gensure, R.H., Yu, X. et al. (2014).
Pilot study of angiogenic response to
yttrium-90 radioembolization with resin micro-
spheres. J Vasc Interv Radiol 25:297–306.
3.10 CONCLUSION Chan, W.S., Poon, W.L., Cho, D.H. et al. (2010).
Transcatheter embolisation of intrahepatic
Vascular considerations are an important part of arteriovenous shunts in patients with hepa-
90Y radioembolization planning and therapy. From tocellular carcinoma. Hong Kong Med J
choosing arterial access to understanding and 16:48–55.
62 Treatment planning part I

Columbo, J.A., Trus, T., Nolan, B. et al. (2015). Kerlan, R.K.J., LaBerge, J.M. (2006). Liver
Contemporary management of median arcu- transplantation: Associated interventions. In
ate ligament syndrome provides early symp- Abrams, H.L., Baum, S.B., Pentecost, M.J.,
tom improvement. J Vasc Surg 62:151–156. (eds.), Abrams’ Angiography: Interventional
Covey, A.M., Brody, L.A., Maluccio, M.A. Radiology, 2nd Edition. Baltimore, MD:
et al. (2002). Variant hepatic arterial anatomy Lippincott Williams & Williams, p. 565.
revisited: Digital subtraction angiogra- Lam, M.G., Banerjee, A., Goris, M.L. et al. (2015).
phy performed in 600 patients. Radiology Fusion dual-tracer SPECT-based hepatic
224:542–547. dosimetry predicts outcome after radio-
Fischman, A.M., Ward, T.J., Patel, R.S. et al. (2014). embolization for a wide range of tumour
Prospective, randomized study of coil embo- cell types. Eur J Nucl Med Mol Imaging
lization versus surefire infusion system during 42:1192–1201.
yttrium-90 radioembolization with resin micro- Lee, A.J., Gomes, A.S., Liu, D.M. et al. (2012). The
spheres. J Vasc Interv Radiol 25:1709–1716. road less traveled: Importance of the lesser
Gaba, R.C., Vanmiddlesworth, K.A. (2012). branches of the celiac axis in liver embolo-
Chemoembolic hepatopulmonary shunt therapy. Radiographics 32:1121–1132.
reduction to allow safe yttrium-90 radioem- Leung, T.W., Lau, W.Y., Ho, S.K. et al. (1995).
bolization lobectomy of hepatocellular carci- Radiation pneumonitis after selective inter-
noma. Cardiovasc Interv Radiol 35:1505–1511. nal radiation treatment with intraarterial
Garin, E., Rolland, Y., Edeline, J. et al. (2015). 90yttrium-microspheres for inoperable
Personalized dosimetry with intensification hepatic tumors. Int J Radiat Oncol Biol Phys
using 90Y-loaded glass microsphere radio- 33:919–924.
embolization induces prolonged overall Liu, D.M., Salem, R., Bui, J.T. et al. (2005).
survival in hepatocellular carcinoma patients Angiographic considerations in patients
with portal vein thrombosis. J Nuclear Med undergoing liver-directed therapy. J Vasc
56:339–346. Interv Radiol 16:911–935.
Graves, P.R., Siddiqui, F., Anscher, M.S. et al. Murata, S., Tajima, H., Nakazawa, K. et al. (2009).
(2010). Radiation pulmonary toxicity: From Initial experience of transcatheter arterial che-
mechanisms to management. Semin Radiat moembolization during portal vein occlusion
Oncol 20:201–207. for unresectable hepatocellular carcinoma
Hamoui, N., Minocha, J., Memon, K. et al. with marked arterioportal shunts. Eur Radiol
(2013b). Prophylactic embolization of the gas- 19:2016–2023.
troduodenal and right gastric arteries is not Pasciak, A.S., McElmurray, J.H., Bourgeois, A.C.
routinely necessary before radioembolization et al. (2015). The impact of an antireflux cath-
with glass microspheres. J Vasc Interv Radiol eter on target volume particulate distribution
24:1743–1745. in liver-directed embolotherapy: A pilot study.
Hamoui, N., Ryu, R.K. (2011). Hepatic radioem- J Vasc Interv Radiol 26:660–669.
bolization complicated by fulminant hepatic Riaz, A., Lewandowski, R.J., Kulik, L.M. et al.
failure. Semin Radiat Oncol 28:246–251. (2009). Complications following radioem-
Hamoui, N., Salem, R., Lewandowski, R.J. (2013a). bolization with yttrium-90 microspheres: A
Embolization of the gastroduodenal and right comprehensive literature review. J Vasc Interv
gastric arteries prior to radioembolization Radiol 20:1121–1130; quiz 1131.
with glass microspheres: Is it always neces- Rose, S.C., Hoh, C.K. (2009). Hepatopulmonary
sary? Abstract # 413. In SIR 2013 38th Annual shunt reduction using chemoembolization to
Scientific Meeting, April 13–18, 2013, New permit yttrium-90 radioembolization. J Vasc
Orleans, LA. Interv Radiol 20:849–851.
Ho, S., Lau, W.Y., Leung, T.W. et al. (1997). Clinical Salem, R., Parikh, P., Atassi, B. et al. (2008).
evaluation of the partition model for estimat- Incidence of radiation pneumonitis after
ing radiation doses from yttrium-90 micro- hepatic intra-arterial radiotherapy with
spheres in the treatment of hepatic cancer. yttrium-90 microspheres assuming uniform
Eur J Nucl Med 24:293–298. lung distribution. Am J Clin Oncol 31:431–438.
References 63

Sangro, B., Gil-Alzugaray, B., Rodriguez, J. et al. Wang, D.S., Louie, J.D., Kothary, N. et al. (2013).
(2008). Liver disease induced by radioembo- Prophylactic topically applied ice to prevent
lization of liver tumors: Description and pos- cutaneous complications of nontarget chemo-
sible risk factors. Cancer 112:1538–1546. embolization and radioembolization. J Vasc
Sergio, A., Cristofori, C., Cardin, R. et al. (2008). Interv Radiol 24:596–600.
Transcatheter arterial chemoembolization Ward, T.J., Tamrazi, A., Lam, M.G. et al. (2015).
(TACE) in hepatocellular carcinoma (HCC): Management of high hepatopulmonary
The role of angiogenesis and invasiveness. shunting in patients undergoing hepatic
Am J Gastroenterol 103:914–921. radioembolization. J Vasc Interv Radiol
Sugano, S., Miyoshi, K., Suzuki, T. et al. (1994). 26:1751–1760.
Intrahepatic arteriovenous shunting due to Welsh, J.S., Kennedy, A.S., Thomadsen, B. (2006).
hepatocellular carcinoma and cirrhosis, and its Selective internal radiation therapy (SIRT)
change by transcatheter arterial embolization. for liver metastases secondary to colorectal
Am J Gastroenterol 89:184–188. adenocarcinoma. Int J Radiat Oncol Biol Phys
Theysohn, J.M., Schlaak, J.F., Muller, S. et al. (2012). 66:S62–S73.
Selective internal radiation therapy of hepato- Xiong, Z.P., Yang, S.R., Liang, Z.Y. et al. (2004).
cellular carcinoma: Potential hepatopulmonary Association between vascular endothe-
shunt reduction after sorafenib administration. lial growth factor and metastasis after
J Vasc Interv Radiol 23:949–952. transcatheter arterial chemoemboliza-
VanDamme, J.P., Bonte, J. (1990). Vascular tion in patients with hepatocellular car-
Anatomy in Abdominal Surgery. New York, cinoma. Hepatobiliary Pancreat Dis Int
NY: Thieme. 3:386–390.
4
Treatment planning part II: Procedure
simulation and prognostication

SHYAM M. SRINIVAS, SANKARAN SHRIKANTHAN, NAICHANG YU, SUSAN


D. KOST, RAM GURAJALA, AND KARUNAKARAVEL KARUPPASAMY

4.1 Background, properties of 4.5.1 LSF evaluated using 99mTc-MAA 76


99mTc-MAA, trends toward 4.5.2 Effect of flow dynamics and
quantification 66 particle size 77
4.1.1 Background—imaging as a 4.5.3 Correlation between
simulation 66 distribution of 99mTc-MAA and
4.1.2 99mTc-MAA physical and absorbed dose 78
biological properties 67 4.5.4 Summary of validity
4.1.3 Trends toward quantification 69 of 99mTc-MAA as a
4.2 Imaging protocol 69 radioembolization surrogate 80
4.2.1 Imaging protocol overview 69 4.6 Alternatives to MAA in procedure
4.2.2 Technical aspects of MAA simulation and/or prognostication 80
injection and imaging 70 4.6.1 Usefulness of preprocedural
4.3 Lung dosimetry 71 CT/MRI in predicting LSF 80
4.3.1 Conventional techniques 71 4.6.2 Usefulness of C-ARM CBCT in
4.3.2 Advanced techniques 72 enhancing safety 81
4.4 Nontarget embolization, radiation 4.6.3 Is radioembolization without
pneumonitis, and their effects 73 99mTc-MAA injection feasible? 81
4.4.1 Extrahepatic uptake and 4.6.4 166Ho microspheres: a potential
clinical strategies used to treat 73 alternative to 99mTc-MAA and
4.4.2 Excessive lung shunting and 90Y radioembolization 82
clinical strategies used to treat 75 4.7 Conclusion 82
4.4.3 Radiation pneumonitis 75 References 83
4.5 Correlation between 99mTc-MAA
and 90Y radioembolization: general
discussion and other considerations 76

65
66 Treatment planning part II

the omission of this very important question of “how


4.1 BaCKGrOUND, PrOPErtIES much” of a cytotoxic agent are we going to give.
OF 99mtC-Maa, trENDS The 90Y radioembolization community is
tOWarD QUaNtIFICatION unique. Its authorized users include radiation
oncologists, nuclear medicine physicians, and
4.1.1 BACKGROUND—IMAGING AS interventional radiologists with the appropriate
A SIMULATION training. As a result, there are those who believe in
precise quantification and simulation, while others
The entire field of radiation oncology is based treat by empirical methods. 90Y radioembolization
on procedure simulation followed by treatment, itself is unsealed, but it is not a systemic injection,
usually with sealed sources. There are abundant which allows more control over the process. It is a
examples in conformal external beam radiation locoregional therapy delivered through the hepatic
therapy, intensity-modulated radiation therapy arterial vasculature with potential for extrahepatic
(IMRT), stereotactic beam radiotherapy (SBRT), shunting, as this chapter further explains. While
gamma knife, and proton beam therapy. In all the 90Y microsphere is handled like a radiophar-
these modalities dose calculations are made based maceutical, it lacks many of the properties of tra-
on pretreatment simulation, followed by treat- ditional radiopharmaceuticals. For example, there
ment of the patient. This process helps to ensure is no metabolism of radioembolic microspheres
that the therapeutic index (i.e., treat disease while as it is an inert permanent implant, resembling a
sparing normal tissue) remains high. With more brachytherapy point source.
sophisticated imaging techniques now readily When glass microspheres (TheraSphere) and
available, simulation is usually based on imag- resin microspheres (SIR-Spheres) were introduced
ing studies such as computed tomography (CT), into clinical usage in the early 2000s, there was
magnetic resonance imaging (MRI), positron no sophisticated mandated simulation technique
emission tomography (PET), and/or ultrasound that would allow for quantification and dosimetry.
(Pereira et al., 2014). Instead, the manufacturer’s guidance simply asked
In nuclear medicine, unsealed source therapy for documentation of lung shunting using planar
is the predominant treatment modality. There are scintigraphy of the radiopharmaceutical 99mTc
examples of near-perfect simulation such as distri- macroaggregated albumin (MAA). 99mTc-MAA is
bution of 123I metascan prior to administering 131I used because of the similar particle size to that of
for ablation in thyroid cancer or the distribution the 90Y microspheres, with the idea that this would
of 99mTc-methylene diphosphonate (MDP) prior be a “poor man’s” simulation of 90Y radioemboli-
to administering 153Sm or 223Ra in prostate can- zation. As we examine the physical properties of
cer (Silberstein et al., 2003). More recently with MAA and 90Y microspheres later in the chapter,
the advent of semiquantitative 68Ga PET imaging, there will be clarity about why MAA is an imper-
simulations are being performed with 68Ga DOTA- fect microsphere surrogate.
d-Phe(1)-Tyr(3)-octreotide (DOTATOC) for neu- With the advent of hybrid single-photon emis-
roendocrine tumors followed by treatment with a sion computed tomography (SPECT)/CT imaging
beta emitter such as 90Y or 177Lu DOTATOC (Baum circa 2005, a new opportunity to perform tomo-
and Kulkarni, 2012). The disconnection between graphic imaging with the potential for quantifi-
radiation therapy using unsealed internal emitters cation has emerged (Beauregard et al., 2011). It
compared with sealed source internal and external has taken nearly 10 years for the field to mature
radiation therapy has been and is the lack of quan- to realize that there is value in performing such
tification. Most radiation oncology treatment plans simulations, and the a priori information from
99mTc-MAA SPECT/CT may finally give the type
have isodose curves planned on the images prior to
the treatment administration, accurately identify- of quantitatively rigorous simulation that is present
ing radiation-absorbed dose to the organs of inter- in the radiation oncology world (Willowson et al.,
est to the nearest Gy. In nuclear medicine, however, 2009, 2011). So while it is an imperfect surrogate at
dosimetry is vague, since many radiotracers have a present, 99mTc-MAA’s potential value is just begin-
multivariable systemic distribution. In the past, the ning to be realized. Of course, there are controver-
lack of quantitative imaging techniques has led to sies as to whether MAA SPECT/CT does actually
4.1 Background, properties of 99mTc-MAA / 4.1.2 99mTc-MAA physical and biological 67

predict the distribution of 90Y microspheres that or the ventilation–perfusion scan typically used
will also be addressed later in this chapter. For now, to evaluate for pulmonary embolism (Neumann
it should be clarified that correlation between MAA et al., 1980). The second indication is to aid in the
and 90Y microspheres has not reached consensus in evaluation of the peritoneovenous (LeVeen) shunt
the literature. While some articles suggest near- patency. This is a rare situation when peritoneal
perfect agreement (Talanow et al., 2010), others are cavity fluid can have systemic circulation entry. The
in disagreement (Wondergem et al., 2013). use of 99mTc-MAA to evaluate for lung shunt frac-
tion and hepatic tumor perfusion with a catheter-
based intra-arterial injection is an off-label usage.
4.1.2 TC-MAA PHYSICAL AND
99m However, this technique has gained widespread
BIOLOGICAL PROPERTIES acceptance and is recommended by both micro-
sphere manufacturers for the calculation of the
Currently, albumin macroaggregates (99mTc-MAA) lung shunt (Package Insert for TheraSphere®, 2014
are supplied by Jubilant DraxImage Inc. and Package Insert for SIR-Sphere®, 2014). This “off-
(Kirkland, Quebec, Canada). The product name label” usage is widely accepted and is also reim-
is “DRAXIMAGE MAA” and it is a kit containing bursed by CMS and U.S.-based insurance carriers.
nonradioactive aggregated human serum albumin The albumin aggregates are sufficiently fragile
(HSA) that can be tagged with 99mTc pertechnetate for the hepatic tumor capillary micro-occlusion to
(MSDS for Draximage®, 2011). be temporary. Erosion and fragmentation reduce
The kit consists of reaction vials that contain the particle size, allowing passage of the aggregates
the sterile, nonradioactive ingredients necessary through the hepatic capillary bed. The fragments
to produce 99mTc albumin-aggregated injection for are then accumulated by the reticuloendothelial
diagnostic use by intravenous injection. Each 10 system. Elimination of the technetium 99mTc aggre-
mL reaction vial contains 2.5 mg of albumin aggre- gated albumin occurs with a half-life of about 2–3 h
gated, 5.0 mg of albumin human, 0.06 mg stannous (Prescribing information for Draximage®, 2010) in
chloride, and 1.2 mg of sodium chloride; the con- lung imaging.
tents are in a lyophilized (freeze-dried) form under A common occurrence in hepatic tumors, espe-
an atmosphere of nitrogen (Prescribing informa- cially hepatocellular carcinoma (HCC), is that of
tion for Draximage®, 2010). patent intrahepatic arteriovenous shunting. The
The aggregated particles are formed by denatur- end result of this shunt is that the 99mTc-MAA par-
ation of human albumin in a heating and aggre- ticles will not be mechanically lodged within the
gation process. Each vial contains 4–8 million tumor vasculature. Instead, they will bypass the
particles. By light microscopy, more than 90% of tumor and become lodged in the next available
the particles are between 10 and 70 µm, while the capillary bed, which is the pulmonary capillary
typical average size is 20–40 µm; none are greater system. Additionally, there are collateral arterial
than 150 µm. No less than 90% of the pertechnetate pathways that can divert the MAA particles to
99mTc added to a reaction vial is bound to aggre- nontargeted capillary beds. Therein lies part of the
gate at preparation time and 99mTc remains bound reason why 99mTc-MAA is useful prior to radioem-
throughout the 6-h lifetime of the preparation bolization. It is beneficial to know if the proposed
(Prescribing information for Draximage®, 2010). catheter position will expose the patient to either
99mTc decays by isomeric transition with a phys- nontarget embolization (NTE) or excessive lung
ical half-life of 6.02 h. Nearly 90% of all disintegra- radiation exposure. The 99mTc-MAA radiotracer,
tions result in the principal photon emission that especially when used with tomographic SPECT/
is useful for detection and imaging studies, which CT imaging, will not only quantify lung shunt,
has a mean energy of 140.5 keV (Prescribing infor- but also identify significant deposition in extrahe-
mation for Draximage®, 2010). patic gastrointestinal (GI) tissues. The implications
The Food and Drug Administration (FDA) of the result of MAA simulation and subsequent
has two currently approved indications and usage strategies to mitigate potential complications will
for 99mTc-MAA: the first most common indica- be discussed in Section 4.4.
tion is as a lung imaging agent which is to evalu- For hepatic imaging, the number of 99mTc-MAA
ate pulmonary perfusion as part of the “V/Q scan” particles per single injection is 200,000–700,000
68 Treatment planning part II

with the suggested number being approximately multiple doses can be obtained from each kit.
350,000—similar to use in lung perfusion study. An individual dosage can range from 2 to 4 mCi
Due to breakdown and subsequent disassociation (74–148 MBq) (Package Insert for TheraSphere®,
of the MAA particles, free 99mTc pertechnetate 2014), but most commonly 4 mCi (148 MBq)
begins to accumulate immediately after injection. dosages are used. Occasionally, hepatic arterial
Therefore, imaging should be performed as soon anatomy may dictate the requirement of 99mTc-
as possible after infusion (Prescribing informa- MAA injection through two distinct catheter
tion for Draximage®, 2010). The physical break- positions for demonstration of the entire liver’s
down of the MAA particles in the liver should shunt potential. Alternatively, the interventional
be longer than in the lung due to lack of motion, radiologist may plan a bilobar radioembolization
leading to a potential for change in relative mea- treatment, which may require infusion at two
sured uptake as the time between injection and distinct catheter positions. To attain reasonable
imaging increases. In addition, the longer the surrogacy with MAA, infusion at the same two
waiting time, the greater the amount of free positions that will be used in subsequent ther-
technetium will be deposited in its expected dis- apy must be performed. In such a situation, two
tribution (salivary and thyroid glands, stomach, separate 2 mCi (74 MBq) 99mTc-MAA dosages are
and kidneys). Since identification of nontarget often prepared in separate syringes so that they
embolization via detection of MAA activity in could be injected from the two desired catheter
stomach is a major utility of the MAA simula- positions.
tion procedure, free 99mTc in the stomach can cer- The effective half-life of 99mTc-MAA can be cal-
tainly be confounding. culated from the formula:
Table 4.1 contrasts the properties of 99mTc-MAA
compared with the two available 90Y radioemboli- 1
= (1 / t physical ) + (1 / t biological )
zation products. It is quite apparent from Table 4.1 teffective
that MAA is close in size to both of the 90Y glass
and 90Y resin products; however, there is an order where t is the half-life.
of magnitude between the number of MAA par- With a physical half-life of 99mTc of 6 h and a
ticles and the number of 90Y glass spheres, and two biological half-life of approximately 2.5 h in the
orders of magnitude between the number of MAA lungs (Prescribing information for Draximage®,
particles and the number of 90Y resin spheres. This 2010), the above formula suggests an effective
has caused much controversy as some believe that half-life of 1.76 h for the lungs. However, this
MAA is not a good surrogate for 90Y while others parameter is purely of academic value. The effec-
do believe it is a useful simulation. tive half-life of 99mTc-MAA in the liver should be
Typically, one MAA kit will be combined with greater given the longer biologic half-life owing
100 mCi (3.7 GBq) of 99mTc pertechnetate so that to lack of mechanical breakdown. In fact, the

Table 4.1 Tc-MAA, an imperfect surrogate


99m

99mtc-Maa Y-90 glass Y-90 resin


Size of particles, mean 20–40 μm (10–70 μm) 20–30 μm (15–35 μm) 30–35 μm (20–60 μm)
(range)
No. of particles, mean 350 k (200–700 k) 4 M for 10 GBq vial 30–60 M (10–80 M)
(range) (1.2–8 M)
Sources: Prescribing information for Draximage®, Available from: http://www.accessdata.fda.gov/drugsatfda_docs/
label/2009/017881s010lbl.pdf, Revised October 2010, Accessed 17 December 2015, 2010; Package Insert
for TheraSphere® Yttrium-90 Glass Microspheres, Available from: http://www.therasphere.com/physicians-
package-insert/TS_PackageInsert_USA_v12.pdf, Revised August 21, 2014, version 12, Accessed 15
December 2015, 2014; NRC Device Registry for Glass Microspheres, NR-0220-D-131-S, Amended August
10, 2015, 2015; Package Insert for SIR-Sphere® Yttrium-90 Microspheres, Available from: http://www.sirtex.
com/media/29845/ssl-us-10.pdf, Revised November 2014, version 10, Accessed 15 December 2015, 2014;
Kennedy, A et al., Int J Radiat Oncol Biol Phys, 68, 13, 2007.
Note: k, thousand; M, million; μm, microns or micrometers.
4.2 Imaging protocol / 4.2.1 Imaging protocol overview 69

biologic half-life in the liver may be closer to 8 h (Amor-Coarasa et al., 2014). If custom kits can
resulting in an effective half-life of 3.4 h for the be created for 68Ga-MAA synthesis, then it is also
liver (Grosser et al., 2016). As time progresses possible to increase the number of MAA particles
following infusion of MAA, the image quality such that it is closer to the number used in the 90Y
degrades and interpretation is confounded by glass or resin radioembolization. This may reduce
the 99mTc-MAA breakdown. The presence of even the potential for discrepancy in distribution due
a small amount of free 99mTc may confuse image to the limited embolic effect associated with tra-
interpretation. Waiting for several hours would ditional MAA. The potential for 68Ga-MAA PET/
be ill advised, as a significant portion of the CT is exceptionally exciting as it is possible to
99mTc would be free, rendering the MAA image run PET/CT scanners with iodinated contrast
suboptimal. and a triphasic liver protocol so that structural
Of interest is a recent article (Grosser et al, 2016) tumor delineation is paired with the MAA distri-
that assessed the biodegradation of 99mTc-MAA ver- bution. In addition, PET/MR may be even more
sus 99mTc HSA. The authors assessed the residual useful as liver tumors can be seen with or with-
activity in MAA and HSA at three time points, 1, 5, out gadolinium contrast and diffusion weighted
and 24 h postinjection, to assess the lung shunt frac- sequences can also provide useful tumor infor-
tion (LSF). As expected, the LSF calculated using mation, although motion artifacts degrage the
MAA changed by 3.9%, 7.7% and 9.9% at the 3 time fusion. There are also newer liver-specific MR
points, respectively, representing a bi-exponential contrast agents such as Eovist®/Primavist® that
decay. Grosser et al. (2016) recommended that have advantages in imaging hepatic lesions like
MAA shunt calculations not be performed based on HCC (Campos et al., 2012). Superimposing the
images greater than 4 h after injection of MAA and MAA distribution over a detailed anatomical
optimally not after 1 h to get an accurate picture of tumor map is a very powerful a priori planning
the shunt fraction. tool, especially if the MAA distribution can be
quantified.

4.1.3 TRENDS TOWARD 4.2 IMaGING PrOtOCOL


QUANTIFICATION
99m Tc-MAA is not the only tracer that can be 4.2.1 IMAGING PROTOCOL
used for simulation in radioembolization. While OVERVIEW
99mTc-MAA is imaged using planar scintigraphy,

SPECT, or SPECT/CT, the use of PET/CT in pre- 99mTc-MAA scintigraphy with angiography for
treatment simulation has sparked some interest. vascular mapping is performed for radioemboliza-
PET is capable of better image quality and more tion treatment planning and detection of potential
accurate quantification compared with SPECT. complications from extrahepatic deposition of 90Y
In some academic centers, the availability of the microspheres. Following infusion of a 99mTc-MAA
68 Ge/68 Ga generator has permitted changing the dosage, planar images of the abdomen and lungs
V/Q scan from a planar/SPECT study to that of a are obtained immediately in anterior and posterior
PET study. Ament et al. has described the use of projections to assess the LSF (American College
a 68Ga tagged aerosol (Galligas) and 68Ga-labeled of Radiology–Society of Interventional Radiology
MAA for the purposes of V/Q scanning (Ament [ACR–SIR] practice parameter for radioemboliza-
et al., 2013). There are various citations in the lit- tion, 2014). Reasonable image acquisition involves
erature that describe the radiochemistry required collecting 140 KeV emissions with a planar gamma
to tag 68Ga to the MAA compound as early as in camera for 2 min using a 20% energy window.
1989 (Even and Green, 1989; Mathias and Green, A planar dual head camera, utilizing both ante-
2008). One recent study describes the use of a rior and posterior heads simultaneously, fulfills
lyophilized kit specifically for creation of the the need for geometric mean (GM) calculation.
PET perfusion agent 68Ga MAA and discusses its Typically, low-energy high resolution (LEHR) or
potential benefit in radioembolization planning equivalent collimators are used for this acquisition.
70 Treatment planning part II

The LSF is calculated using the GM of ante- 4.2.2 TECHNICAL ASPECTS OF


rior and posterior projections. Regions of interest MAA INJECTION AND
(ROIs) are drawn around the whole lung and the IMAGING
whole liver in the anterior and posterior projec-
tions and respective counts are obtained. The LSF In metastatic liver disease, MAA injection can
is then determined using the following formulae: be performed in the proper hepatic artery, due to
the relatively lower incidence of high lung shunt-
ing compared with patients with HCC. HCC can
GM =  ROIAnterior × ROIPosterior (4.1)
be associated with portal vein thrombosis, and
an increased prevalence for direct arteriovenous
shunting bypassing the capillary bed, increas-
GM Lung
Lung shunt fraction  (LSF) =   (4.2) ing the risk of radiation pneumonitis (RP). In the
GM Liver + GM Lung author’s experience, significant LSFs (>20%) are
seen only in cases of HCC. Among 109 patients who
Example contours are illustrated in Figure 4.1a received preradioembolization MAA infusion and
and b for a patient that has a large (~18 cm) HCC imaging over the past year at the author’s institu-
and an LSF of 18%. A coronal hepatic protocol tion, there were six cases with LSF > 20%, all with
CT scan of this patient is shown in Figure 4.1c for HCC. Five of the six also had portal vein throm-
comparison. Total count values from lung fields or bosis. These data are representative of the modern
the liver field in anterior and posterior views are worldwide experience with radioembolization.
used to calculate GM and the LSF (Equations 4.1 In patients with bilobar HCC, without gross
and 4.2). vascular shunting into the hepatic or portal vein,
99mTc-MAA can be injected into the proper hepatic

artery. However, if shunting is seen, it is often pref-


Anterior Posterior erable to do a unilobar injection of MAA to assess
one lobe at a time. A follow-up MAA scan can be
performed separately, prior to treatment of the
Lung ROI other lobe. In certain situations of variant arte-
rial anatomy such as replaced right hepatic artery,
fractionated dosages of 99mTc-MAA are injected to
cover the entire liver in one sitting if possible, 2–3
mCi of MAA in the replaced right hepatic, and the
remaining 2–3 mCi into the left hepatic artery.
Liver ROI As previously mentioned, it is preferable to assess
(a) (b) LSF and imaging as soon as possible after the MAA
injection. While 99mTc pertechnetate can confound
interpretation of NTE to GI tissue, it can also result
in an overestimation of LSF. Compton scattering
also artificially increases the LSF and, therefore, the
calculated lung dose. The lung dose is important, as
a radiation dose >30 Gray (Gy) given to the lungs in
one treatment or a cumulative dose >50 Gy in mul-
(c)
tiple treatments is considered a relative contraindi-
cation (Salem and Thurston, 2006a).
Figure 4.1 Calculation of lung shunt fraction Planar scintigraphy is usually employed to cal-
in a patient following injection of 4.0 mCi of
99mTc-MAA into the right hepatic artery. Contours culate LSF. However, Yu et al. (2013) described a
are drawn to identify counts in the lungs and liver new method of calculating the mean lung dose
on anterior (a) and posterior (b) projections. A (MLD) for radioembolization of liver cancer based
coronal reformat of a pretreatment hepatic com- on 99mTc-MAA SPECT/CT, which can provide
puted tomography scan of the patient’s tumor is a more accurate estimate of radiation risk to the
shown in (c). lungs. In addition, SPECT and SPECT/CT also have
4.3 Lung dosimetry / 4.3.1 Conventional techniques 71

increased sensitivity of detecting extrahepatic NTE hepatic distribution. The follow-up 18FDG-PET/CT
compared with planar imaging. Ahmadzadehfar scan (Figure 4.2c) about 3 months posttreatment
et al. (2010) reported the sensitivity for detect- shows complete response with resolution of the pre-
ing extrahepatic deposition increased from 32% viously present focal 18FDG uptake on the baseline
with planar imaging and 41% with SPECT alone PET scan.
to 100% with SPECT/CT. The specificity for NTE
detection was 98% with planar and SPECT imag- 4.3 LUNG DOSIMEtrY
ing and 93% with SPECT/CT. SPECT/CT imaging
is able to detect extrahepatic activity predicting
sites at risk for NTE and alter treatment planning 4.3.1 CONVENTIONAL
in about 29% of cases. Identification of these cases TECHNIQUES
following MAA simulation allows for corrective
actions included coiling the responsible vessels, Treatment of hepatic cancer with radioemboliza-
repositioning the infusion catheter, or in some tion relies on administering a high concentration
cases cancellation of procedure (Ahmadzadehfar of 90Y microspheres to the tumor. Judicious place-
et al., 2010). Examples are shown in Section 4.4. ment of the injection catheter and relative enhance-
In addition to pretreatment identification of pos- ment of the tumor vasculature make it possible to
sible NTE and LSF, the pattern of distribution of deliver high doses to the tumor, while controlling
MAA activity in the SPECT/CT also helps in the the amount of activity to normal liver. Due to the
prediction of posttreatment response (Garin et al., pathological nature of the tumor vasculature, arte-
2012). Figure 4.2a shows the 18F-fluorodeoxyglucose riovenous shunting of the injected microspheres may
(18FDG)-PET/CT of a patient with a tumor in the be significant, leading to the deposition of shunted
posterior right hepatic lobe, with the corresponding microspheres to the lung and risk of radiation-
MAA scan (Figure 4.2b) demonstrating matching induced pneumonitis. This risk is controlled through
prequalification of patients for the treatment. As pre-
viously mentioned, mean lung doses (MLDs) from
shunting greater than 30 Gy from a single treatment
or cumulative MLD >50 Gy is a contraindication to
radioembolization (Riaz et al., 2014).
The LSF can be estimated from pretreatment
99mTc-MAA scintigraphy, as described in Equations

4.1 and 4.2. Conventionally, the LSF is determined


(a) (b) using the partition model (Ho et al., 1996) from
the number of counts in the lungs (MAA lung) and
liver (MAA liver) designated by regions on a two-
dimensional (2D) planar image as previously indi-
cated. Using the 99mTc-MAA activity distribution
as a surrogate for the activity distribution of radio-
(c) active microspheres and assuming that all micro-
spheres become trapped in capillaries and decay
Figure 4.2 The pattern of distribution of macro-
with rate of the physical half-life only (i.e., there is
aggregated albumin (MAA) activity in the single- no biological removal), the medical internal radia-
photon emission computed tomography (SPECT)/ tion dose (MIRD) formalism can be used to esti-
CT helps in predicting posttreatment response. mate the absorbed dose to the lungs. The MIRD
(a) Fluorodeoxyglucose (FDG) avid tumor in the formalism (Loevinger and Berman, 1976) assumes
posterior right lobe (18FDG-PET/CT), with the cor- local energy deposition whereby the absorbed dose
responding MAA scan (b) demonstrating match- to a volume of mass M with 90Y activity A is given
ing activity. (c) Follow-up 18FDG-PET/CT scan as follows (Ho et al., 1996):
(about 3 months posttreatment) shows complete
response with resolution of the previously present
A ( GBq ) × 49.98(J ⋅ s)
focal FDG uptake on the baseline positron emis- Davg ( Gy ) =       (4.3)
sion tomography (PET) scan. M (kg)
72 Treatment planning part II

Lung dose is determined using the total image acquisition may result in misregistration of
injected activity Atotal and the LSF by A lung = Atotal activity around the diaphragm. Liver activity can
× LSF with an assumed mass of 1 kg [International be blurred into the lung region defined by the CT
Commission on Radiological Protection (ICRP), that was acquired within a few seconds during a
1975]. Patient-specific evaluation of lung mass is specific phase of the respiratory cycle, resulting in
not routinely performed for clinical radioembo- an overestimation of the lung activity.
lization procedures owing to the lack of volumet- Yu et al. (2013) proposed a method of calcu-
ric information provided by planar scintigraphy. lating the MLD based on 99mTc-MAA SPECT/
This extension for calculating the absorbed dose CT. MLD is approximated by the mean dose in
to the lung is given in Equation 4.4 and can be a subregion of the lung (Lungsub) that excludes
applied to routine radioembolization treatment the portion of lung that is within 2 cm of the
scenarios: diaphragm in order to avoid spillover from liver.
LSFs to Lungsub were determined from activities
Atotal ( GBq ) × LSF × 49.98(J ⋅ s) in Lungsub and external body within the scan
Davg ( Gy ) =       (4.4) region contoured on the CT. MLD was deter-
1(kg)
mined retrospectively for 71 patients by Equation
While convenient for routine clinical care, esti- 4.3 using a mass calculated from the volume of
mating lung dose from a 2D projection image has Lungsub and an assumed lung density of 0.3 g/cm3
several limitations. Without an anatomical image, (Van Dyk et al., 1982). The lung doses calculated
lung and liver contours on planar scintigraphy can from SPECT were compared with those from an
be subjective. Because of the high uptake of activ- LSF estimated from lung and liver regions on pla-
ity in the liver, small variations in the definition of nar scintigraphy assuming a total lung mass of
the border between the liver and lung may result 1000 g. In almost all patients, the lung dose from
in large variations in the measured activity to the planar scintigraphy exceeded the dose derived
lung. In addition, planar scintigraphy is not eas- from SPECT, with a mean planar scintigraphy to
ily corrected for photon attenuation and scatter, SPECT lung dose ratio of 3.8 ± 4.0. For patients
leading to large potential uncertainties in lung and at highest risk for radiation-induced pneumonitis
liver MAA activity. Using the GM of LSFs deter- from treatment with estimated lung doses greater
mined from a conjugate pair of anterior and pos- than 15 Gy according to planar scintigraphy, the
terior planar images may mitigate the differences ratio was 2.7 ± 1.1. The authors further studied
in photon attenuation due to depth; however, the the effect of ignoring the misregistration due to
GM technique does not correct for scatter and fails breathing and failure to make attenuation and
to compensate for photon attenuation differences scattering corrections during the reconstruc-
due to varying tissue densities as exhibited in liver tion of the SPECT images, concluding that each
and lung. will result in roughly 50% overestimation of
the lung dose. Recognizing the subjectivity and
4.3.2 ADVANCED TECHNIQUES potential inaccuracy of LSF estimation based on
planar scintigraphy, the authors recommended
The use of 99mTc-MAA SPECT can improve the using SPECT for more accurate mean lung dose
estimation of lung dose from arteriovenous shunt- estimation.
ing. Three-dimensional (3D) image reconstruc- A recent study by Kao et al. (2014) further
tion in conjunction with attenuation and scatter demonstrated the uncertainties using planar
corrections using tissue densities derived from a images for estimating MLD and the importance
coregistered anatomical CT image improves the of using SPECT/CT for individualized MLD esti-
quantification of liver and lung activity uptake for mation. Kao et al. (2014) compared lung doses
calculation of the LSF. The anatomical CT image calculated from 99mTc-MAA SPECT/CT to the
also provides information about the lung volume conventional planar scintigraphy method for a
and the ability to perform patient-specific den- cohort of 30 Southeast Asian patients. The tech-
sitovolumetry. However, the time to obtain the nique for estimating lung dose from 3D SPECT/
necessary planar projections for 3D SPECT recon- CT was similar to that used by Yu et al. (2013).
struction is lengthy and free breathing during the LSFs were determined using counts from lung
4.4 NTE, RP, and their effects / 4.4.1 Extrahepatic uptake and clinical strategies used to treat 73

and liver volumes segmented on the CT. A dis-


tance of 1.5 cm superior to the domes of the dia- 4.4 NONtarGEt EMBOLIZatION,
phragm was selected to exclude lung counts from raDIatION PENUMONItIS,
misregistration of activity in the liver due to free aND tHEIr EFFECtS
breathing during image acquisition. MLD was
determined using Equation 4.3 with a patient- Planar 99mTc-MAA imaging is performed to
specific lung parenchyma mass determined by determine LSF and to exclude gastrointestinal
CT densitovolumetry for the lung volume above shunting. In particular, 99mTc-MAA SPECT/CT
the exclusion zone. Lung and liver activities were imaging provides fused functional and anatomic
also measured from conventional planar scin- data, adds clarity and confidence in the identifi-
tigraphy and a statistically significant difference cation of extra hepatic flow, and can guide target
between mean LSFs from planar (5.96 ± 4.59%) embolization of extrahepatic feeders. However,
and SPECT (7.36 ± 4.96%) was found. However, SPECT/CT should not be solely used to abso-
the mean lung absorbed doses from both methods lutely exclude gastrointestinal shunting. Rather,
was not statistically significant. The mean lung it should be considered an adjunctive imaging
mass for their patient cohort was 830 g, not vastly modality of the gastrointestinal tract. Exclusion
different from the standard lung mass of 1000 g. of gastrointestinal flow should be accomplished
For patients with particularly smaller lung mass, by use of the combined information obtained
such as patients postlobectomy, the planar image from hepatic angiography, 3D CT angiography,
method may underestimate the MLD. and SPECT imaging.
RP from arteriovenous shunting is a rare
complication in treatment of liver cancer using
90Y radioembolization, with an occurrence of less
4.4.1 EXTRAHEPATIC UPTAKE AND
than 1% (Chan et al., 1995; Salem et al., 2008). A CLINICAL STRATEGIES USED
contraindication of treatment arises only if lung TO TREAT
dose is greater than a single treatment dose of
30 Gy or cumulative dose of 50 Gy (Riaz et al., As discussed in Chapter 3, planning angiography
2014). The conventional method of determining is necessary before radioembolization as it pro-
lung and liver activity from planar scintigraphy vides an overview of normal and variant anatomy.
of 99mTc-MAA may result in an overestimation With advanced technologies in C-arm CT and
of MLD due to the inaccuracy of delineating new catheter types, the frequency of prerequisite
regions representing lung and liver volumes and prophylactic coil embolization to prevent hepati-
the inability to apply attenuation and scatter cor- coeneteric flow has decreased. Nontarget deposi-
rections to 2D images. Likewise, the necessity of tion of microspheres can have grave consequences.
using an assumed standard lung mass of 1000 g The most common sites of undesired particle
for dosimetric calculation from planar scintigra- deposition are in the gall bladder, gastrointestinal
phy may contribute to an over- or underestima- (GI) system, and pulmonary system.
tion of patient lung dose depending on patient Radiation cholecystitis results from uptake of
size and lung physiology. Lung dose estimation radioactive microspheres in the gall bladder. This
from 3D 99mTc-MAA SPECT/CT using patient- can be prevented by preemptive identification of
specific lung mass offers an improved and more the cystic artery and placement of the catheter
precise assessment of lung dose from shunting. tip beyond the origin. If blood flow is significant
However, even if SPECT/CT is used, interpreta- and injection beyond the origin is not possible,
tion of lung dose from 90Y microsphere activity then embolization may be considered (Salem and
based on surrogate 99mTc-MAA scintigraphic Thurston, 2006b). This is managed by supportive
imaging is still associated with error. Erosion and care and if refractory, cholecystectomy should be
fragmentation of the albumin aggregates reduce performed (Atassi, 2008). 99mTc-MAA simula-
the particle size resulting in a larger amount of tion provides a sensitive predictor for radiation
99mTc-MAA activity traveling to the lungs com- cholecystitis. Figure 4.3 shows uptake in the gall-
pared with 90Y microspheres, which have a rigid bladder along with the uptake in the liver and the
structure. tumor in the 99mTc-MAA SPECT/CT. This example
74 Treatment planning part II

illustrates the importance of 99mTc-MAA simula- dynamics during infusion (Murthy et al., 2007).
tion in order to prevent complications such as radi- These patients should be aggressively managed
ation cholecystitis. with proton pump inhibitors to prevent more
The incidence of GI ulceration is less than serious complications such as ulceration and/or
5% if meticulous techniques are used (Mallach perforation that may eventually require surgical
et al., 2008; Szyszko et al., 2007). Although pre- management.
treatment hepatic angiography should assist in Figure 4.4 shows gastric uptake in the pylo-
reducing these complications, misdirected micro- rus identified on 99mTc-MAA SPECT/CT that
spheres can enter hepaticoenteric flow. These could have potentially resulted in ulceration.
patients may have intense embolic pain during Prophylaxis included coiling the culprit gastric
or after the procedure. As shown in Chapter 13, artery. Figure 4.5a shows the planning angio-
if GI NTE is suspected, confirmation using 90Y gram following routine occlusion of the gastro-
PET/CT after the microsphere infusion can aid in duodenal artery. The MAA uptake detailed in
medical management of these patients. Definitive Figure 4.4 prompted additional occlusion of the
diagnosis can be made by upper endoscopy. These right-gastric artery shown in Figure 4.5b. This
toxicities can be attributed to unrecognized vari- prophylactic measure allowed for safe adminis-
ants, collateral circulation, and changes in flow tration of 90Y radioembolization.
Inadvertent delivery of microspheres can also
occur through the vessels supplying the anterior
abdominal wall via the falciform artery (Liu et al.,
2005) resulting in radiation dermatitis (Leong et
al., 2009). These side effects can be avoided by pro-
phylactic embolization during pretreatment angi-
ography (Meyer et al., 2014). At our institute, we
use an ice pack/saline bag placed over the abdomi-
nal wall that is presumed to cause vasospasm and
redirect flow into the intrahepatic circulation
(Wang, 2013). Figure 4.6 shows the 99mTc-MAA
SPECT/CT of a patient with HCC. There is MAA
uptake corresponding to the tumor in the right
lobe of liver. Additionally, there is a linear focus
(a)
of activity extending inferiorly into the anterior

(b)

Figure 4.3 Uptake in the gallbladder (arrow) from


shunting into the cystic artery, with the risk of Figure 4.4 99mTc-MAA SPECT/CT image shows
radiation-induced cholecystitis. (a) Hepatic pro- potential activity in the pylorus. Additional pro-
tocol CT and (b) 99mTc-MAA SPECT/CT following phylactic steps must be taken prior to treatment
infusion of 4.0 mCi of 99mTc-MAA. to avoid gastrointestinal (GI) complication.
4.4 NTE, RP, and their effects / 4.4.3 Radiation pneumonitis 75

(a) (b)

Figure 4.5 Angiography before and after 99mTc-MAA SPECT/CT. (a) Planning angiogram following
routine occlusion of the gastroduodenal artery. (b) After identification of nontarget embolization
(NTE) to the pylorus on MAA SPECT/CT, coil occlusion of the right-gastric artery was performed.

4.4.2 EXCESSIVE LUNG SHUNTING


AND CLINICAL STRATEGIES
USED TO TREAT
Arteriovenous anastomoses or shunts in the liver
parenchyma or tumor cause the lung shunting
that potentially could result in RP after radioem-
bolization (Wright et al., 2012; Leung et al., 1995).
NTE
Various strategies for managing excessive LSF are
described in the literature. These include cancel-
lation of the procedure in any patient with mark-
edly elevated hepatopulmonary shunting (LSF >
20%) (Leung et al., 1995), reduction in the micro-
sphere dose (10% < LSF < 20%) (Elschot et  al.,
2011), bland embolization, or chemoemboliza-
Figure 4.6 Abdominal wall shunting. 99mTc-MAA tion of the shunt (Gaba and Vanmiddlesworth,
SPECT/CT in a patient with hepatocellular carci- 2012) and balloon occlusion of the hepatic vein
noma. MAA uptake corresponding to the tumor while delivering the microspheres could allow
is present in the right lobe of liver. A sagittal safe 90Y delivery (Ward et  al., 2015). Additional
reformat shows linear activity extending inferiorly techniques are discussed in other chapters of this
into the anterior abdominal wall in the region of book.
the umbilicus, representing extrahepatic shunt-
ing via the falciform artery.
4.4.3 RADIATION PNEUMONITIS
abdominal wall in the region of the umbilicus, rep-
resenting potential extrahepatic NTE via the fal- RP represents an acute manifestation of radiation-
ciform artery. Due to pretreatment identification induced lung disease caused by increased hepato-
using MAA, the aforementioned ice pack prophy- pulmonary shunting with an associated increased
laxis was employed during the treatment of this estimated radiation dose to the lungs. Most of the
patient preventing side effects. cases in the literature are a result of external beam
76 Treatment planning part II

irradiation after treatment for lung and breast can- an objective response was seen. However, when
cers (Leung et al., 1995), lymphoma, and whole-body given prophylactically they failed to prevent RP.
irradiation for stem cell transplantation (Camus, Pentoxifylline (a platelet inhibitor with immuno-
2004). MLD, lung volume receiving a specified dose, modulating/anti-inflammatory properties medi-
and normal tissue complication probability (NTCP) ated through interleukin-1/tumor necrosis factor)
are the three widely studied parameters used to is thought to be helpful in preventing radiotoxic-
assess the risk for RP (Graves et al., 2010). Rodrigues ity by inhibiting platelet aggregation and tumor
et al. (2004) highlighted that direct comparisons necrosis factor (Ozturk et al., 2004).
between studies could not be achieved given the In summary, it should be emphasized that the
heterogeneity of outcome variables. However, most incidence of RP is low. A cautious approach should
studies did show an association between dose–vol- be of paramount importance when the hepatopul-
ume histogram parameters and RP. It is noted that monary shunt fraction would result in a lung dose
dosimetric parameters play a lesser role than patient exceeding 30 Gy (Murthy et al., 2005).
characteristics for the prediction of lung toxicity
(Dehing-Oberije et al., 2009). Ramella et al. (2010)
determined addition of ipsilateral constraints (i.e., 4.5 COrrELatION BEtWEEN
99mtC-Maa aND 90Y
volume of lung receiving 30 Gy) to standard lung
dosimetric factors in patients with RP in non–small- raDIOEMBOLIZatION:
cell lung cancer treated with 3D conformal RT and GENEraL DISCUSSION aND
concurrent chemotherapy resulted in a reduction in OtHEr CONSIDEratIONS
the incidence of pneumonitis from 14.4% to 6.8%.
According to Riaz et al. (2009), RP is a complica-
In the current practice of radioembolization, esti-
tion that has not been studied optimally. In their
mating the LSF plays a key role in preventing inad-
report involving 58 patients, none developed RP
vertent RP. As previously discussed, determination
with cumulative lung doses exceeding 50 Gy (Salem
of LSF involves the use of 99mTc-MAA as a surrogate
et al., 2008). However, there is a case report (Wright
for radioembolization in a separate planning pro-
et al., 2012) with RP in a patient with a lung dose
cedure. The distribution of these particles within
of 31.0 ± 13.0 Gy. It is presumed in this case that
the liver, however, is in large part currently ignored
RP was not predicted using the currently used 90Y
in routine clinical practice. Although 99mTc-MAA
dosimetry models that assume uniform distribution
is trusted as a surrogate for 90Y microspheres in the
in the lungs (Salem et al., 2008), which may explain
measurement of the LSF, its utility in the accurate
the findings of Wright et al. (2012).
modeling of hepatic distribution of radioemboliza-
RP patients generally present with gradual onset
tion has not been unequivocally demonstrated. In
of dyspnea, fever, bronchoalveolar lymphocytosis
this section, literature reviewing the accuracy of
and eosinophilia. This initially presents as a mild 99mTc-MAA as a surrogate for radioembolization
restrictive process on pulmonary testing, with ill-
is reviewed, beginning first with evaluation of LSF
defined patchy opacities and ground-glass nodu-
with the use of MAA.
larity in a symmetric (i.e., “bat-wing”) pattern with
relative peripheral/hilar sparing 1–2 months after
therapy. The features can also resemble an organiz- 4.5.1 LSF EVALUATED USING
ing or chronic eosinophilic pneumonia. These may 99mTC-MAA
resolve or progress toward localized fibrosis, trac-
tion bronchiectasis, and focal honeycombing. Late It is routine practice to inject 99mTc-MAA into the
complications include pneumothorax and super- hepatic arterial branch 2–4 weeks before the injec-
infections (Leung et al., 1995; Camus, 2004; Riaz tion of 90Y microspheres. After injection of 99mTc-
et al., 2009). MAA, planar scintigraphy is routinely performed
The first line of management of RP is corticoste- and ROIs over the lungs and the liver are used
roids, which may reduce the degree of inflamma- to measure LSF as previously described. In addi-
tion (Leung et al., 1995). Rubin and Casarett (1968) tion, SPECT/CT imaging of the upper abdomen is
demonstrated that when corticosteroids were employed at many sites to visually assess the dis-
given after clinical pneumonitis had developed, tribution of particles in the liver and extrahepatic
4.5 Correlation between 99mTc-MAA and 90Y radioembolization/ 4.5.2 Effect of flow dynamics 77

territory to ensure future safe delivery of 90Y radio- on rats, the mean tumor to liver arterial perfusion
active microspheres. ratio (T:N) was 3:1 for 15- and 32.5-μm spheres but
LSF is known to be higher for HCC than for other 1:1 for 50-μm microspheres (Van de Wiele et al.,
tumors (8.0% vs. 6.3%; p = .048) (Olorunsola et al., 2012). Clearly, the size of the particles plays a key
2015). In one study, high LSF (>20%) occurred in role in their distribution within the liver and sub-
14% of HCC cases but in only 3% of other tumors sequent shunting away from the liver.
(p = .004) (Gaba et al., 2014). Colorectal cancer Two types of microspheres are currently avail-
(CRC) metastases (median LSF, 10.6%) and HCC able to perform radioembolization for the treat-
(11.7%) are known to have a significantly larger ment of liver cancer: glass-based and resin-based
LSF than metastases from breast cancer (7.4%; microspheres. Resin 90Y microspheres measure
p < .005) (Powerski et al., 2015). Similar results approximately 32.5 ± 2.5 μm in size, whereas the
are reported through the literature, and we have glass-based 90Y microspheres measure approxi-
noticed consistency at our institution as well in mately 25 ± 5 μm. In addition, the total number
a retrospective review of 39 patients who under- of spheres per GBq is approximately 20 million
went right lobe 99mTc-MAA injections (37 lobar, 2 for 90Y resin microspheres and only approximately
segmental) before radioembolization for HCC at 400,000 for 90Y glass microspheres (Cremonesi
the Cleveland Clinic. Among these patients, the et al., 2014). Thus, glass microspheres have a signif-
mean 99mTc-MAA LSF was 5.9% (SD, 0.03%; range, icantly higher amount of radioactivity per micro-
0.5–12.1%), with no significant difference in LSF sphere. As a result, for a similar radiation dose
among patients with (n = 7) or without (n = 32) 90Y resin microspheres are expected to be more

extrahepatic distribution of 99mTc-MAA. embolic than 90Y glass microspheres; however, this
In a study by Lambert et al. (2010), low-quality depends on the prescribed radiation dose and the
whole-body scintigraphy images (defined as visu- size of the vascular bed in the liver that is to be
alization of the kidneys when adequate scaling of treated.
the whole-body scintigraphy image had to be per- Compared with 90Y microspheres, 99mTc-MAA
formed to assess the liver) were correlated with a particles infused in the planning stage have a
higher LSF. The authors found that 14% of the 90 wider range of sizes (5–100 μm), with 80%–90%
studies assessed were considered to be of low qual- of the particles falling within the range of 10–70
ity and suggested that LSF was overestimated in μm (Table 4.1, Zophel et al., 2009). The 99mTc-
this group. MAA particles undergo enzymatic hydrolysis
In the vast majority of patients with primary or and are phagocytized by reticuloendothelial cells.
secondary hepatic tumors, LSF measured by 99mTc- As opposed to 90Y microspheres, the radioactiv-
MAA is less than 20%. A small fraction of HCC ity associated with a 99mTc-MAA dosage does not
tumors is known to be associated with higher LSF necessary vary linearly with particle number. For
(Refaat and Hassan, 2014). In addition, quality of example, doubling the particle size will increase
the scintigraphy images has a significant impact on the average radioactivity per particle by a factor of
the measurement of LSF and a low-quality study 4. Therefore, a particle with a diameter of 40 μm
may result in overestimation of LSF. will contain 16 times more radioactivity than a
particle with a diameter of 10 μm (Van de Wiele
et al., 2012).
4.5.2 EFFECT OF FLOW DYNAMICS The heterogeneous composition of small
AND PARTICLE SIZE (<20  μm) and large (>60 μm) particles with sig-
nificantly different radiation doses per particle
Arterioles feeding liver metastases in humans within a dose of 99mTc-MAA is likely to affect
average 30–40 μm in diameter. When flowing imaged intrahepatic and extrahepatic distribution
through arteries, particles concentrate in a peri- and have an effect on shunt quantification imag-
arteriolar fashion. Hence, the concentration of ing, since small particles are more likely to pass
particles entering a side branch will be lower than through the hepatic capillary bed. This effect may
the concentration in the main channel. In addi- combine with the propensity of planar scintigra-
tion, smaller particles tend to reach the periphery phy to overestimate LSF due to scatter and attenu-
of the liver, whereas larger ones do not. In a study ation, previously discussed. In a study involving 23
78 Treatment planning part II

patients with primary and secondary liver malig- However, in a study of 17 patients (14 with
nancies, 99mTc-MAA scans were found to signifi- HCC, 3 with CRC), Ho et al. (1996) found good
cantly overestimate LSF when compared with gold correlation between the doses estimated using the
standard postradioembolization 90Y PET/CT scans partition model based on T:N and intraoperative
(6% vs. 1.8%; p < .01) (Song et al., 2015). In spite dosimetry in tumors (r = 0.862) and background
of the more heterogeneous composition in a vial, liver (r = 0.804). Ho et al. determined the T:N using
99mTc-MAA is still used universally as a simulation 99mTc-MAA by dividing the average count rates of

surrogate for 90Y radioembolization. the tumor by the average count rates of the normal
liver. This ratio can be used to estimate the activ-
ity of 90Y microspheres that would be partitioned
4.5.3 CORRELATION BETWEEN between the tumor and the normal liver compart-
DISTRIBUTION OF 99mTC-MAA ment. If the activity delivered to the tumor can
AND ABSORBED DOSE be estimated, Equation 4.3 can then be employed
to determine the amount of 90Y microspheres
In clinical practice, the distribution of 99mTc-MAA required to achieve a certain tumoricidal dose
particles is expected to be similar to the distribu- or to keep below a tolerance limit of normal liver
tion of 90Y microspheres, allowing the particles to tissue. Additional details on the utilization of the
serve as a surrogate for the microspheres. However, partition model for hepatic dosimetry are provided
in several studies, the reliability of 99mTc-MAA as in Chapter 5. Unfortunately, this technique is not
a surrogate has been questioned. When prescrib- commonly utilized in routine clinical practice.
ing the radiation dosage, one should understand As additional conflicting evidence regarding
the significant differences in the characteristics of the validity of 99mTc-MAA as a radioembolization
99mTc-MAA and 90Y resin or glass microspheres, surrogate is discussed below, one should keep in
including the size range of the particles/micro- mind that clinical measurement of T:N is likely
spheres and the total number of particles/micro- to vary substantially from patient to patient. This
spheres delivered. Presuming that 99mTc-MAA and variation is not necessarily indicative of inaccuracy
90Y microspheres are delivered at the same site of or error and has been shown in large patient stud-
infusion in the liver, one can still expect a differ- ies (Ilhan et al., 2015a). In general, higher values
ence in their distribution due to differences in flow of T:N occur in cases of neuroendocrine tumors,
kinetics. The physical properties of the injected HCC, and cholangiocellular carcinoma, while
agent and blood flow pattern from the tip of the lower T:N commonly occurs in cases of mammary
catheter at the moment of infusion will dictate the cancer, CRC, and sarcoma.
distribution kinetics of infused particles. This may
explain why procedures are rarely cancelled fol-
lowing suboptimal hepatic distribution of 99mTc- 4.5.3.1 Data suggesting the
MAA obtained in simulation. validity of 99mTc-MAA as a
A study cohort of 66 patients with a total of 435 radioembolization surrogate
colorectal liver metastases showed that response to
90Y resin microspheres was independent of qualita-
Ilhan et al. (2015b) compared the pattern of uptake
tive grading on the degree of pretreatment 99mTc- in different liver tumors obtained using 99mTc-MAA
MAA uptake in the tumor. Hence, patients could SPECT with that of 90Y bremsstrahlung SPECT fol-
not be excluded from radioembolization based on lowing radioembolization using 90Y resin micro-
99mTc-MAA distribution (Ulrich et al., 2013). In
spheres. Among a cohort of 502 patients, 20% had
response to subsequent questions raised about the primary hepatic tumors (HCC, 12%; cholangiocel-
possibility of catheter position being an important lular carcinoma, 8%) and the remaining patients
factor in these results, the authors later reported had metastases from several different primary
additional results of a subgroup analysis in which tumors. The 99mTc-MAA and 90Y bremsstrahlung
the catheter tip was placed in an identical position images were coregistered with contrast-enhanced
for both 99mTc-MAA and 90Y microspheres (41 of CT or MR images. Analysis demonstrated that
the original 66 patients); there was a similar lack of lesions with high uptake on 99mTc-MAA SPECT
correlation (p > .05) (Amthauer et al., 2014). also had high uptake of 90Y microspheres. The
4.5 Correlation between 99mTc-MAA and 90Y radioembolization/ 4.5.3 Correlation between distribution 79

correlation between 99mTc-MAA SPECT and 90Y determined to non-target liver were not statisti-
microsphere uptake was significant but weak (r cally different.
= 0.26; p < .001) (Ilhan et al., 2015b). Other authors For some of the discrepancies reported in
have performed similar analyses comparing 99mTc- the distribution of 99mTc-MAA particles and 90Y
MAA SPECT to posttreatment 90Y bremsstrahlung microspheres, differences in the exact location
SPECT with findings suggesting reasonable agree- of the catheter tip at the time of delivery of these
ment (Knesaurek et al., 2010). materials may have played a role. This hypothesis
In Section 4.5.3.2, several reports suggesting was examined by Jiang et al. (2012) by review-
poor agreement between 99mTc-MAA and radio- ing the perfusion differences between 81 paired
embolization will be reviewed. However, it is 99mTc-MAA hepatic SPECT and posttherapy 90Y

important to note that agreement of spatial dis- bremsstrahlung SPECT studies; corresponding
tribution may not be necessary for 99mTc-MAA to angiograms were also reviewed. When the catheter
serve as a valid tool for predictive hepatic dosim- tip was placed in proximity to an arterial bifurca-
etry using the partition model. For example, Kao tion or a small branch, this seemed to alter micro-
et al. (2013) compared established 90Y PET/CT sphere perfusion or trajectory and was found to be
to pretreatment 99mTc-MAA SPECT/CT in 23 associated with mismatch (Jiang et al., 2012).
patients treated using resin microspheres. Using
posttreatment 90Y PET/CT as the gold standard,
dosimetry based on MAA SPECT showed good 4.5.3.3 Other limitations of
agreement with a median relative error of just 99mTc-MAA simulation
3.8% (max = 13.2%).
Although the anatomical distribution of 99mTc-
MAA particles is considered a surrogate for the
4.5.3.2 Data suggesting 99mTc-MAA distribution of 90Y microspheres, this technique
is a poor radioembolization fails to quantify the functional aspect of the liver.
surrogate Lam et al. (2015) studied the role of intra-arterial
injection of 99mTc-labeled sulfur colloid (SC),
Several examples from the literature have which was injected after 99mTc-MAA-SPECT
reported a lack of correlation in the distribution in the same procedure as a biomarker for func-
of 99mTc-MAA and 90Y radiomicrospheres within tional liver. The authors used the combined
the liver. In a study to assess the ability of 99mTc- information to study voxel-based partitioning
MAA to predict 90Y distribution in 39 patients and dosimetry for subsequent 90Y radioemboli-
treated using 90Y resin microspheres, the pre- zation using resin microspheres in 98 patients
dicted amount of 90Y activity in Couinaud liver and glass microspheres in 24 patients. Through
segments based on 99mTc-MAA SPECT was com- a fusion of 99mTc-MAA SPECT and 99mTc-SC
pared with the actual amount of 90Y based on images, the liver was divided into four compart-
90Y bremsstrahlung SPECT. The absolute mean ments based on uptake (+) and lack of uptake
difference between the estimated and actual 90Y (–) of each tracer: tumor (99mTc-MAA+, SC–);
absorbed dose was around 30 Gy, and a difference irradiated functional liver (99mTc-MAA+, SC+);
of more than 30% of the mean activity per mil- nonirradiated functional liver (99mTc-MAA–,
liliter was found in 32% of the 225 segments ana- SC+); and tumor necrosis, cysts, and major ves-
lyzed (Wondergem et al., 2013). sels (99mTc-MAA–, SC–). Independent of the
While data presented by Kao et al. (2013) sup- type of microspheres used, HCC had a higher
ported the accuracy of MAA-based tumor predic- median tumor/median functional liver absorbed
tive dosimetry using 90Y PET/CT as a gold standard, dose ratio than other tumor types (median 1.8;
Song et al. (2015) have reported some discordance p = .02). The median tumor absorbed dose was
in a 30 patient cohort. Tumor-absorbed dose esti- correlated with response in both univariate and
mated using 99mTc-MAA SPECT/CT was found to multivariate analyses, and the maximum change
be significantly lower than that estimated using in toxicity grade from baseline after radioem-
90Y PET/CT (135.4 ± 64.2 Gy vs. 185.0 ± 87.8 Gy; bolization was associated with the absorbed
p <  .01). However, differences in absorbed dose dose in functional liver tissue (p < .05). With
80 Treatment planning part II

these results, Lam et al. (2015) demonstrated the may also be considered as a potential method to
potential use of 99mTc-SC as a tracer that could improve concordance with radioembolization.
individualize the tolerance of background liver However, such a practice might result in decreased
in each patient, thereby allowing clinicians to T:N during treatment due to the pre-embolic effect
adjust the radioembolization dose to maximize of MAA if treatment is performed before MAA
tumor response while minimizing the risk of has been completely cleared. Unfortunately, the
radioembolization-induced liver disease. time for complete clearance to occur, particularly
in the setting of highly variable neoplasm absent
4.5.4 SUMMARY OF VALIDITY of Kupffer cells, is not known. MAA retention is
known to be prolonged when there is a reduction
OF 99mTc-MAA AS A
in the number of Kupffer cells (Tanaka et al., 1996;
RADIOEMBOLIZATION Rimola et al., 1984 ; Bilzer et al., 2006).
SURROGATE
Differences between tracer distributions are likely
due to the differences between 99mTc-MAA par- 4.6 aLtErNatIVES tO Maa IN
ticles and 90Y microspheres. The number of 90Y PrOCEDUrE SIMULatION
microspheres usually delivered is several orders aND/Or PrOGNOStICatION
of magnitude higher than the number of 99mTc-
MAA particles delivered, which likely results in
4.6.1 USEFULNESS OF
an embolization effect and increases redistribution
into background liver, reducing the correlation in
PREPROCEDURAL CT/MRI IN
measured T:N. Such a difference might be higher PREDICTING LSF
with resin microspheres than with glass micro- Previous studies have suggested that pretreatment
spheres due to the vast differences in the number hepatic protocol CT may have a role in prognosti-
of microspheres within a comparable prescribed cation of response to radioembolization in patients
radiation dose (Table 4.1). In addition, catheter tip with HCC. Tumor hypervascularity compared
position and changes in tumor vascularity (due to with background liver and the amount of intra-
tumor growth or histological changes) between the tumoral blood flow estimated on CT has been
planning and treatment angiograms might also reported to correlate with disease response. In an
play a key role in the differences between 99mTc- analysis of CT scans performed before and after 90Y
MAA particle and 90Y microsphere distribution. glass microsphere radioembolization in 23 patients
Based on the results of several studies, we can with unresectable HCC, prolonged progression-
safely conclude that the distribution and tumor free survival was associated with lower LSF, higher
uptake of 99mTc-MAA particles, which are currently central tumor hypervascularity, and well-defined
used as a surrogate agent, does not consistently tumor margins, whereas shorter progression-free
demonstrate equivalence with the distribution and survival was associated with abutment of the por-
uptake of the therapeutic agent (90Y microspheres). tal vein by the tumor (Salem et al., 2013).
The LSF measured by 99mTc-MAA might be overes- Morsbach et al. (2013) prospectively evaluated
timated in some patients and it is therefore possible the ability of CT perfusion to predict morpho-
that some patients may be unnecessarily excluded logic response and survival in 38 patients with
from radioembolization. Hence, 99mTc-MAA in liver metastases who subsequently underwent 90Y
its current form is not an ideal surrogate for 90Y resin microsphere radioembolization; dose was
radioembolization. calculated using the BSA method. Five seconds
To minimize this discordance, a tighter fil- after contrast material injection (50 mL of iopro-
tration of 99mTc-MAA particles to sizes that mide), 12 spiral acquisitions covering the liver were
more closely match the size range of 90Y micro- obtained in the 4D spiral mode. Arterial perfusion
spheres might be considered. In addition, efforts (AP) in target liver lesions was significantly higher
to increase the embolic burden of 99mTc-MAA in the responders than in the nonresponders (37.5
to more closely approximate 90Y microspheres vs. 11.8 mL/min; p < .001). A cutoff AP of 16 mL
4.6 Alternatives to MAA in procedure / 4.6.3 Is radioembolization without 99mTc-MAA 81

per 100 mL/min had a sensitivity of 100% and a identification of extrahepatic shunting and paren-
specificity of 89% for predicting therapy response. chymal enhancement in radioembolization, van
In a single-center retrospective study of 70 den Hoven et al. (2016) conducted a prospective
patients with HCC, infiltrative morphologic development study. The authors found that the
structure, tumor burden greater than 50%, por- variable contrast and scan delay determined using
tal vein invasion, and arterioportal shunting timing parenchymal enhancement on digital sub-
were significantly associated with high (>20%) traction angiography were more effective than the
LSF in multivariate analysis (Gaba et al., 2014). contrast and scan delay determined with a proto-
Similarly, in a study using pretreatment multi- col that used either a fixed 6-s delay and 10-s scan
phase CT, strong tumor contrast enhancement or a 5-s low-dose scan setting applied to reduce
was found to be associated with a significantly breathing artifacts in combination with a variable
larger LSF than in tumors with little enhance- delay (van den Hoven et al., 2016).
ment (11.7% vs. 8.3%; p < .001). In addition, In the future, multispin/multiphase CBCT may
patients with compression (LSF = 13.9%) or be used during planning to identify features asso-
tumor thrombosis (15.8%) of a major portal ciated with safe and favorable clinical outcomes
vein branch had a significantly higher LSF than and to aid in modifying required 90Y dose activ-
patients with a normal portal vein (8.1%) (both p ity. CBCT may also be used to predict high LSF
< .001) (Powerski et al., 2015). and tumor enhancement characteristics that are
Finally, in a multivariate analysis of findings unlikely to produce desired outcomes, thus allow-
on CT (n = 134) or MRI (n = 18) among patients ing for modification of the treatment plan to other
with primary and secondary hepatic tumors, early forms of embolization in the same session.
hepatic vein opacification and hepatic vein tumor
thrombus or occlusion were associated with a sig-
nificantly higher LSF. Sensitivity and specificity of 4.6.3 IS RADIOEMBOLIZATION
early hepatic vein opacification originating from WITHOUT 99mTC-MAA
the tumor were 78% and 93%, respectively (positive INJECTION FEASIBLE?
likelihood ratio, 10.5), for predicting high (>20%)
LSF (Olorunsola et al., 2015). In a major proportion of patients who undergo
radioembolization, LSF is estimated to be sig-
nificantly less than 20%. As described in Sections
4.6.2 USEFULNESS OF C-ARM CBCT 4.6.1 and 4.6.2, there are several findings on CT/
IN ENHANCING SAFETY MRI and pretreatment angiography that are asso-
ciated with a high lung shunt. In addition, CBCT
With the ability of modern angiographic units to is increasingly used during planning angiogra-
acquire C-arm cone beam CT (CBCT) images, phy, which has improved our ability to prevent
multiplanar evaluation of the tumor, background inadvertent extrahepatic uptake in the abdomen,
liver, and extrahepatic enhancement can now be thus reducing our reliance on 99mTc-MAA SPECT
evaluated during planning angiography (Pellerin imaging. Hence, obtaining such information
et al., 2013). This technique has demonstrated before injection of 99mTc-MAA might help clini-
increased sensitivity in the detection of extrahe- cians to predict which patients are likely to have
patic enhancement when compared with digital a high LSF.
subtraction angiography or 99mTc-MAA imaging Among the most recent 39 patients at The
in a small cohort (Louie et al., 2009). A larger study Cleveland Clinic to undergo treatment planning
evaluated the utility of pretreatment CBCT to cor- using 99mTc-MAA injection for right lobe HCC, the
rectly identify the presence of extrahepatic NTE. highest evaluate LSF was 12.1%. At this maximum
This effort found that CBCT prior to radioembo- lung shunt, a radioembolization absorbed dose of
lization was associated with a negative predictive 120 Gy to a typical right liver lobe measuring 1000
value for extrahepatic shunting of 95% (van den cc in volume would result in 17.1 Gy to the lungs,
Hoven et al., 2016). Despite these positive findings, well within aforementioned safety limits. Using a
there is no single standard C-arm CBCT protocol predetermined target liver volume, a single-session
in use. In an attempt to optimize a protocol for planning angiogram immediately followed by 90Y
82 Treatment planning part II

microsphere infusion without 99mTc-MAA infusion (median, 5.5 Gy; p < .001) (Elschot et al., 2014).
might be feasible. However, such a practice would These results call into question the validity of our
require further validation of methods to prognos- long-standing practice of relying on 99mTc-MAA
ticate LSF without simulating using 99mTc-MAA. estimates to predict risk of RP.
There is already some active effort to condense These data are not mentioned merely to further
radioembolization therapy into a single-session illustrate potential shortcomings associated with
treatment. For example, in a recent study, glass simulation using 99mTc-MAA, but also a potential
microsphere radioembolization was performed on benefit of 166Ho radioembolization. Much of this
14 patients by combining the 99mTc-MAA infusion, chapter has been devoted to analyzing differences
LSF assessment, and injection of 90Y microspheres in the physical properties of 90Y radioemboliza-
on the same day (Gates et al., 2014). It is possible tion and 99mTc-MAA as a source of error. However,
that such examples are a first step into more aggres- since 166Ho radioembolization treatment plan-
sive investigation into the possibility of radioem- ning is based on infusion of a scout dose of 166Ho
bolization treatment on selected patients without microspheres, the agent used for both simulation
scintigraphic evaluation of LSF. and therapy are identical aside from the number
of microspheres infused (Prince et al., 2013). Use
of 166Ho radioembolization, therefore, has the
4.6.4 HO MICROSPHERES: A
166
potential to minimize the mismatches currently
POTENTIAL ALTERNATIVE reported between the distribution and activity of
TO 99mTC-MAA AND 90Y the diagnostic tracer (99mTc-MAA) and therapeutic
RADIOEMBOLIZATION tracer (90Y glass or resin microspheres).

As discussed in Chapter 1, radioactive


Holmium-166 (166Ho) microspheres are seen as
a possible alternative to 90Y microspheres. 166Ho
microspheres emit high-energy beta-radiation,
4.7 CONCLUSION
which is used for therapeutic purposes, as well as
gamma-radiation, which allows for direct scintig- 99m Tc-MAA is the standard of care for preradio-
raphy. In addition, Ho is a highly paramagnetic embolization simulation for the evaluation of
element that can be visualized effectively on MRI. LSF to prevent RP. 99mTc-MAA SPECT/CT can
This allows measurement of radioactivity using also aide in the prognostication of extrahepatic
diagnostic scintigraphy, which can then be used NTE, allowing for appropriate prophylaxis and
for accurate therapeutic dosimetry and MRI-based avoidance of treatment-related side effects. In
high-resolution imaging of the biodistribution of addition, pretreatment 99mTc-MAA SPECT/CT is
the tracer. also used as a treatment planning tool by provid-
In a prospective clinical study in 14 patients ing tumor and normal liver predictive dosimetry.
with unresectable liver metastases treated with Unfortunately, due primarily to physical differ-
166 Ho microsphere radioembolization, the accu- ences between 99mTc-MAA and 90Y microspheres,
racy of lung-absorbed dose estimates by 99mTc- MAA is an imperfect surrogate for radioembo-
MAA-based diagnostic imaging was compared lization and the accuracy with which it fulfills
with the accuracy of estimates directly measured any of the aforementioned functions is limited.
following radioembolization using 166Ho SPECT/ While alternatives to 99mTc-MAA are available
CT. Lung-absorbed doses were significantly which may provide superior accuracy, such as
166 Ho microspheres, there are many other issues
overestimated by 99mTc-MAA imaging (Smits
et al., 2012). In addition, as expected, the actual to consider before moving away from an estab-
absorbed dose in the lungs was better predicted by lished and effective treatment protocol currently
diagnostic 166Ho microsphere SPECT/CT imag- under worldwide clinical use. Instead, the radio-
ing (median, 0.02 Gy) than by 166Ho microsphere embolization treatment team will be well served
planar scintigraphy (median, 10.4 Gy; p < .001), by carefully understanding and considering the
99mTc-MAA SPECT/CT imaging (median, 2.5 Gy; limitations of MAA in the medical management
p < .001), and 99mTc-MAA planar scintigraphy of patients.
References 83

REFERENCES nasopharyngeal carcinoma. Int J Radiat Oncol


Biol Phys 33(3):569–577.
ACR–SIR practice parameter for radioemboliza- Cremonesi, M. et al. (2014). Radioembolization of
tion with microsphere brachytherapy device hepatic lesions from a radiobiology and dosi-
(RMBD) for treatment of liver malignancies: metric perspective. Front Oncol 4:210.
Revised 2014 (resolution 17). Dehing-Oberije, C. et al. (2009). The importance
Ahmadzadehfar, H. et al. (2010). The significance of patient characteristics for the prediction
of 99mTc-MAA SPECT/CT liver perfusion imag- of radiation-induced lung toxicity. Radiother
ing in treatment planning for 90Y-microsphere Oncol 91:421–426.
selective internal radiation treatment. J Nucl Elschot, M. et al. (2011). Quantitative evaluation
Med 51:1206–1212. of scintillation camera imaging characteristics
Ament, S. et al. (2013). PET lung ventilation/per- of isotopes used in liver radioembolization.
fusion imaging using (68)Ga aerosol (Galligas) PLoS One 6(11):e26174.
and (68)Ga-labeled macroaggregated albu- Elschot, M. et al. (2014). (99m)Tc-MAA over-
min. Recent Results Cancer Res 194:395–423. estimates the absorbed dose to the
Amor-Coarasa, A. et al. (2014). Lyophilized kit for lungs in radioembolization: A quantita-
the preparation of the PET perfusion agent tive evaluation in patients treated with
[68Ga]-MAA. Int J Mol Imaging 2014:1–7. 166 Ho-microspheres. Eur J Nucl Med Mol

Amthauer, H., Ulrich, G., Grosser, O., Imaging 41(10):1965–1975.


Ricke, J. (2014). Reply: Pretreatment dosim- Even, G., Green, M. (1989). Gallium-68-labeled
etry in HCC radioembolization with (90)Y macroaggregated human serum albu-
glass microspheres cannot be invalidated min, 68Ga-MAA. Int J Rad Appl Instrum
with a bare visual evaluation of (99m) B 16(3):319–121.
Tc-MAA uptake of colorectal metastases Gaba, R., Vanmiddlesworth, K. (2012).
treated with resin microspheres. J Nucl Med Chemoembolic hepatopulmonary shunt reduc-
55(7):1216–1218. tion to allow safe yttrium-90 radioemboliza-
Atassi, B. et al. (2008). Biliary sequelae following tion lobectomy of hepatocellular carcinoma.
radioembolization with Yttrium-90 micro- Cardiovasc Intervent Radiol 35 (6):1505–1511.
spheres. J Vasc Interv Radiol 19(5):691–697. Gaba, R. et al. (2012). Characteristics of pri-
Baum, R., Kulkarni, H. (2012). THERANOSTICS: mary and secondary hepatic malignancies
From molecular imaging using Ga-68 labeled associated with hepatopulmonary shunting.
tracers and PET/CT to personalized radionu- Radiology 271(2):602–612.
clide therapy—The Bad Berka Experience. Garin, E. et al. (2012). Dosimetry based on
Theranostics 2(5):4375–447. 99mTc-macroaggregated albumin SPECT/
Beauregard, J. et al. (2011). Quantitative 177Lu CT accurately predicts tumor response and
SPECT (QSPECT) imaging using a commer- survival in hepatocellular carcinoma patients
cially available SPECT/CT system. Cancer treated with 90Y-loaded glass microspheres:
Imaging 11(1):56–66. Preliminary results. J Nucl Med 53(2):255–263.
Bilzer, M., Roggel, F., Gerbes, A. (2006). Role of Gates, V. et al. (2014). Outpatient single-session
Kupffer cells in host defense and liver disease. yttrium-90 glass microsphere radioemboliza-
Liver Int 26(10):1175–1186. tion. J Vasc Interv Radiol 25(2):266–270.
Campos, J., Sirlin, C., Choi, J. (2012). Graves, P., Siddiqui, F., Anscher, M., Movsas,
Focal hepatic lesions in Gd-EOB-DTPA B. (2010). Radiation pulmonary toxicity: From
enhanced MRI: The atlas. Insights Imaging mechanisms to management. Semin Radiat
3(5):451–474. Oncol 20:201–207.
Camus, P., Kudoh, S., Ebina, M. (2004) Interstitial Grosser, O. et al. (2016). Pharmacokinetics of
lung disease associated with drug therapy. 99mTc-MAA and 99mTc-HSA microspheres used

Br J Cancer 91(suppl 2):S18–S23. in pre-radioembolization dosimetry: Influence


Chan, A. et al. (1995). A prospective random- on the liver-lung shunt. J Nucl Med 57(2).
ized study of chemotherapy adjunctive Published February 9, 2016 as doi:10.2967/
to definitive radiotherapy in advanced jnumed.115.169987
84 Treatment planning part II

Ho, S. et al. (1996). Partition model for estimating of tumour cell types. Eur J Nucl Med Mol
radiation doses from yttrium-90 microspheres Imaging 42(8):1192–1201.
in treating hepatic tumours. Eur J Nucl Med Lambert, B. et al. (2010). 99mTc-labelled mac-
23(8):947–952. roaggregated albumin (MAA) scintigraphy
Ilhan, H. et al. (2015a). Systematic evaluation for planning treatment with 90Y micro-
of tumoral 99mTc-MAA uptake using spheres. Eur J Nucl Med Mol Imaging
SPECT and SPECT/CT in 502 patients 37(12):2328–2333.
before 90Y radioembolization. J Nucl Med Leong, Q. et al. (2009). Radiation dermatitis fol-
56(3):333–338. lowing radioembolization for hepatocellular
Ilhan, H. et al. (2015b). Predictive value of 99mTc- carcinoma: A case for prophylactic emboliza-
MAA SPECT for 90Y-labeled resin micro- tion of a patent falciform artery. J Vasc Interv
sphere distribution in radioembolization of Radiol 20:833–836.
primary and secondary hepatic tumors. J Nucl Leung, T., Lau, W., Ho, S. (1995). Radiation
Med 56(11):1654–1660. pneumonitis after selective internal radiation
International Commission on Radiological treatment with intraarterial 90yttrium-micro-
Protection. (1975). Reference Man: spheres for inoperable hepatic tumors. Int J
Anatomical Physiological and Metabolic Radiat Oncol Biol Phys 33:919–924.
Characteristics. ICRP Publication 23. Oxford: Liu, D. et al. (2005). Angiographic considerations
Pergamon Press. in patients undergoing liver-directed therapy.
Jiang, M. et al. (2012). Segmental perfusion dif- J Vasc Interv Radiol 16:911–935.
ferences on paired Tc-99m macroaggregated Loevinger, R., Berman, M. (1976). A Revised
albumin (MAA) hepatic perfusion imaging Schema for Absorbed-Dose Calculations for
and yttrium-90 (Y-90) Bremsstrahlung imag- Biologically Distributed Radionuclides. MIRD
ing studies in SIR-sphere radioembolization: Pamphlet 1 Biologic. New York, NY: Society of
Associations with angiography. J Nucl Med Nuclear Medicine.
Radiat Ther 3:122. Louie, J. et al. (2009). Incorporating cone-beam
Kao, Y. et al. (2014). Personalized predic- CT into the treatment planning for yttrium-90
tive lung dosimetry by technetium-99m radioembolization. J Vasc Interv Radiol
macroaggregated albumin SPECT/CT for 20(5):606–613.
yttrium-90 radioembolization. EJNMMI Res Mallach, S. et al. (2008). An uncommon cause
4(1):33. of gastro-duodenal ulceration. World J
Kao, Y. et al. (2013). Post-radioembolization Gastroenterol 14:2593–2595.
yttrium-90 PET/CT—part 2: Dose–response Material Safety Data Sheet for DRAXIMAGE®-
and tumor predictive dosimetry for resin MAA. (2011). Available from: http://www.
microspheres. EJNMMI Res 3(57):1–12. draximage.com/data/PDF/51_en.pdf.
Kennedy, A. et al. (2007). Recommendations Jubilant DraxImage Inc., revised July 2011, revi-
for radioembolization of hepatic malignan- sion no. 4. Accessed 15 December 2015.
cies using yttrium-90 microsphere brachy- Mathias, C., Green, M. (2008). A convenient route
therapy: A consensus panel report from The to [Ga-68]Ga-MAA for use as a particulate
Radioembolization Brachytherapy Oncology PET perfusion tracer. Appl Radiat Isotopes
Consortium. Int J Radiat Oncol Biol Phys 66(12):1910–1912.
68:13–23. Meyer, C. et al. (2014). Feasibility of temporary
Knesaurek, K. et al. (2010). Quantitative com- protective embolization of normal liver tissue
parison of yttrium-90 (90Y)-microspheres and using degradable starch microspheres during
technetium-99m (99mTc)-macroaggregated radioembolization of liver tumours. Eur J Nucl
albumin SPECT images for planning 90Y Med Mol Imaging 41:231–237.
therapy of liver cancer. Tech Cancer Res Morsbach, F. et al. (2013). Computed tomo-
Treatm 9(3):253–261. graphic perfusion imaging for the prediction
Lam, M. et al. (2015). Fusion dual-tracer SPECT- of response and survival to transarterial radio-
based hepatic dosimetry predicts outcome embolization of liver metastases. Invest Radiol
after radioembolization for a wide range 48(11):787–794.
References 85

Murthy, R. et al. (2005). Yttrium-90 microsphere Prince, J. et al. (2013). Holmium-166 micro-
therapy for hepatic malignancy: Devices, indi- spheres for image-guided radioemboliza-
cations, technical considerations, and poten- tion: No need for patient isolation after
tial complications. Radiographics 25(Suppl treatment [abstract]. J Vasc Interv Radiol
1):S41–S55. 24(4):S157–S158.
Murthy, R. et al. (2007). Gastrointestinal com- Ramella, S. et al. (2010) Adding ipsilateral V20
plications associated with hepatic arterial and V30 to conventional dosimetric con-
yttrium-90 microsphere therapy. J Vasc Interv straints predicts radiation pneumonitis in
Radiol. 18:553–561. stage IIIA-B NSCLC treated with combined-
Neumann, R., Sostman, H., Gottschalk, A. (1980). modality therapy. Int J Radiat Oncol Biol
Current status of ventilation-perfusion imag- Phys 76:110–115.
ing. Semin Nucl Med 10(3):198–217. Refaat, R., Hassan, M. (2014). The relationship
NRC Device Registry for Glass Microspheres. between the percentage of lung shunting on
(2015). NR-0220-D-131-S, Amended August Tc-99m macroaggregated albumin (Tc-99m
10, 2015. MAA) scan and the grade of hepatocellular
Olorunsola, O. et al. (2015). Imaging predic- carcinoma vascularity. Egypt J Radiol Nucl
tors of elevated lung shunt fraction in Med 45(2):333–342.
patients being considered for yttrium-90 Riaz, A. et al. (2009). Complications following
radioembolization. J Vasc Interv Radiol radioembolization with yttrium-90 micro-
26(10):1472–1478. spheres: A comprehensive literature review. J
Ozturk, B., Egehan, I., Atavci, S., Kitapci, Vasc Interv Radiol 20:1121–1130.
M. (2004). Pentoxifylline in prevention of Riaz, A., Rafia, A., Riad, S. (2014). Side effects
radiation-induced lung toxicity in patients of yttrium-90 radioembolization. Frontiers
with breast and lung cancer: A double-blind Oncol. 4. (198):1–11
randomized trial. Int J Radiat Oncol Biol Phys Rimola, A. et al. (1984). Reticuloendothelial
58:213–219. system phagocytic activity in cirrhosis and its
Package Insert for TheraSphere® Yttrium-90 relation to bacterial infections and prognosis.
Glass Microspheres. (2014). Available from: Hepatology 4(1):53–58.
http://www.therasphere.com/physicians- Rodrigues, G. et al. (2004). Prediction of radia-
package-insert/TS_PackageInsert_USA_v12. tion pneumonitis by dose–volume histogram
pdf. Revised August 21, 2014, version 12. parameters in lung cancer—A systematic
Accessed 15 December 2015. review. Radiother Oncol 71:127–138.
Package Insert for SIR-Sphere® Yttrium-90 Rubin, P., Casarett, G. (1968). Clinical Radiation
Microspheres. (2014). Available from: http:// Pathology. Philadelphia, PA: W.B. Saunders.
www.sirtex.com/media/29845/ssl-us-10.pdf. Salem, M. et al. (2013). Radiographic parameters
Revised Nov 2014, version 10. Accessed 15 in predicting outcome of patients with hepa-
December 2015. tocellular carcinoma treated with yttrium-90
Pellerin, O. et al. (2013). Can C-arm cone-beam microsphere radioembolization. ISRN Oncol
CT detect a micro-embolic effect after 2013:538376.
TheraSphere radioembolization of neuroen- Salem, R. et al. (2008). Incidence of radiation
docrine and carcinoid liver metastasis? Cancer pneumonitis after hepatic intra-arterial radio-
Biother Radiopharm 28(6):459–465. therapy with yttrium-90 microspheres assum-
Pereira, G., Traughber, M., Muzic, R. (2014). The ing uniform lung distribution. Am J Clin Oncol
role of imaging in radiation therapy planning: 31(5):431–438.
Past, present, and future. BioMed Res Int Salem, R., Thurston, K. (2006a). Radioembolization
2014:1–9. with 90-yttrium microspheres: A state-of-the-
Powerski, M. et al. (2015). Hepatopulmonary art brachytherapy treatment for primary and
shunting in patients with primary and second- secondary liver malignancies: Part 1—Technical
ary liver tumors scheduled for radioemboliza- and methodologic considerations. J Vasc Interv
tion. Eur J Radiol 84(2):201–207. Radiol 17:1251–1278.
86 Treatment planning part II

Salem, R., Thurston, K. (2006b). Radioembolization Van den Hoven, A. et al. (2016). Use of C-arm
with 90-yttrium microspheres: A state-of-the- cone beam CT during hepatic radioemboliza-
art brachytherapy treatment for primary and tion: Protocol optimization for extrahepatic
secondary liver malignancies: Part 2: Special shunting and parenchymal enhancement.
topics. J Vasc Interv Radiol 17:1425–1439. Cardiovasc Intervent Radiol 39(1):64–73.
Silberstein, E. et al. (2003). Society of nuclear Van Dyk, J., Keane, T., Rider, W. (1982). Lung den-
medicine procedure guideline for palliative sity as measured by computerized tomogra-
treatment of painful bone metastases version phy: Implications for radiotherapy. Int J Radiat
3.0. Approved January 25, 2003. Available Oncol Biol Phys 8:1363–1372.
from: http://interactive.snm.org/docs/pg_ Wang, D. et al. (2013). Prophylactic topically
ch25_0403.pdf. Accessed 31 January 2016. applied ice to prevent cutaneous complica-
Smits, M. et al. (2012). Holmium-166 radioemboli- tions of nontarget chemoembolization and
sation in patients with unresectable, chemo- radioembolization. J Vasc Interv Radiol 24(4):
refractory liver metastases (HEPAR trial): A 596–600.
phase 1, dose-escalation study. Lancet Oncol Ward, T. et al. (2015). Management of high hepa-
13(10):1025–1034. topulmonary shunting in patients undergo-
Song, Y. et al. (2015). PET/CT-based dosimetry in ing hepatic radioembolization. J Vasc Interv
90Y-microsphere selective internal radiation Radiol 26(12):1751–1760.
therapy: Single cohort comparison with pre- Willowson, K., Bailey, D., Baldock, C. (2009).
treatment planning on (99m)Tc-MAA imag- Quantitative analysis of [Tc-99m]-MAA lung
ing and correlation with treatment efficacy. uptake in the clinical work-up for the treat-
Medicine (Baltimore) 94(23):e945. ment of liver tumours with [Y-90]-SirSpheres.
Szyszko, T. et al. (2007). Management and pre- J Nucl Med 50 (suppl 2):1438.
vention of adverse effects related to treat- Willowson, K., Bailey, D., Baldock, C. (2011).
ment of liver tumours with 90Y microspheres. Quantifying lung shunting during plan-
Nucl Med Commun 28:21–24. ning for radio-embolization. Phys Med Biol
Talanow, R. et al. (1996). Ability of pre-therapy 56(13):N145.
Tc99m MAA SPECT/CT to predict the dis- Wondergem, M. et al. (2013). 99mTc-macroaggre-
tribution of Y90 radiomicrosphere therapy gated albumin poorly predicts the intrahe-
as defined by Bremsstrahlung SPECT/CT. patic distribution of 90Y resin microspheres
Abstract and poster presented at RSNA in hepatic radioembolization. J Nucl Med
annual meeting in 2010. 54(8):1294–1301.
Tanaka, M. et al. (1996). Pathomorphological Wright, C. et al. (2012). Radiation pneumonitis
study of Kupffer cells in hepatocellular carci- following yttrium-90 radioembolization: Case
noma and hyperplastic nodular lesions in the report and literature review. J Vasc Interv
liver. Hepatology 24(4):807–812. Radiol. 23:669–674.
Ulrich, G. et al. (2013). Predictive value of Yu, N. et al. (2013). Lung dose calculation with
intratumoral 99mTc-macroaggregated SPECT/CT for 90Yttrium radioembolization
albumin uptake in patients with colorectal of liver cancer. Int J Radiat Oncol Biol Phys
liver metastases scheduled for radioembo- 85(3):834–859.
lization with 90Y-microspheres. J Nucl Med Zophel, K., Bacher-Stier, C., Pinkert, J., Kropp,
54(4):516–522. J. (2009). Ventilation/perfusion lung scin-
Van de Wiele, C. et al. (2012). SIRT of liver metas- tigraphy: What is still needed? A review
tases: Physiological and pathophysiological considering technetium-99m-labeled macro-
considerations. Eur J Nucl Med Mol Imaging aggregates of albumin. Ann Nucl Med
39(10):1646–1655. 23(1):1–16.
5
Treatment planning part III: Dosimetric
considerations in radioembolization
with glass and resin microspheres

ALEXANDER S. PASCIAK, AUSTIN C. BOURGEOIS, AND YONG C. BRADLEY

5.1 Background 87 5.8.1 The empiric model for resin


5.2 Absorbed dose, effective dose, and microsphere treatment planning 97
dosage 88 5.8.2 The BSA model for resin
5.2.1 Absorbed dose 89 microsphere treatment planning 98
5.2.2 Dosage 89 5.8.3 Glass microsphere treatment
5.2.3 Effective dose 89 planning 98
5.3 One microsphere or millions of 5.8.4 The partition model 99
microspheres? 89 5.8.5 Comparing treatment planning
5.4 Distribution in tissue 91 models 99
5.5 Calculating the absorbed dose 93 5.9 Contouring liver and tumor volumes 100
5.6 Tumor and normal liver endpoints 94 5.9.1 Automatic and semiautomatic
5.6.1 Normal liver endpoints 94 segmentation 101
5.6.2 Tumor endpoints 95 5.9.2 Manual segmentation 101
5.7 Tumor-to-normal uptake ratio 95 5.10 Treatment planning calculations 103
5.7.1 Using MAA as an estimation 5.10.1 Lobar therapy 103
tool 96 5.10.2 Selective therapy 106
5.7.2 Other tools? 97 5.11 Conclusions 109
5.8 Treatment planning strategies 97 References 109

However, differences between the two products


5.1 BaCKGrOUND extend beyond microsphere composition and
include the method in which yttrium-90 (90Y) is
As mentioned in Chapter 1, the most widely bound to each microsphere, the presence of other
used radioembolization products worldwide are radioactive contaminants, and, most importantly,
SIRTeX SIR-Spheres (SIRTeX Technology Pty, Lane the activity of 90Y per sphere. Treatment planning
Cove, Australia) and BTG Theraspheres (BTG for the two products differs in some ways due to the
International Ltd., London, United Kingdom), often activity per microsphere, as discussed at a microdo-
referred to as resin and glass microspheres, respec- simetric level in Chapter 9. However, the primary
tively, in reference to their material composition. differences in treatment planning methodologies

87
88 Treatment planning part III

discussed later in this chapter may be related more


to the use of legacy planning models, which have 5.2 aBSOrBED DOSE,
shown utility in tens of thousands of treatments. EFFECtIVE DOSE, aND
Some of the differences between the two primary DOSaGE
radioembolization products were introduced in
Chapter 1 but have also been restated in Table 5.1. Ariel and Padula (1978a, 1978b) reported the first
It should be noted that for resin microspheres, cases of the clinical use of 90Y microspheres in the
the projected lower-limit activity per microsphere intra-arterial treatment of colorectal metastases to
listed in Table 5.1 is based on the shelf-life of the the liver. Ariel combined intra-arterial infusion of
product as determined by the manufacturer. At resin 90Y microspheres with chemotherapy in the
the end of the 15-day shelf-life of the glass product form of 5-fluorouracil. Ariel’s patients received rel-
(Giammarile et al., 2011), the low-end activity per atively large dosages of 90Y, ranging from 100 to 150
microsphere is approximately 50 Bq rather than the mCi (3.7–5.5 GBq) that lead to a robust response in
commonly referenced lower limit of 300 Bq. After their cohort of 65 patients. This method involved
90Y microsphere delivery using both percutaneous
15 days, the total activity in the largest 20 GBq vial
of glass microspheres would be far less than 1 GBq, injection and intra-arterial approaches via a sur-
gically placed hepatic arterial catheter. Since this
which is insufficient for an effective treatment. The
initial experience, intra-arterial 90Y microsphere
use of glass microspheres approaching their expi-
therapy has significantly evolved in both treatment
ration date is consequentially impractical in many
and manufacturing processes.
situations. Despite the now widespread use of radioembo-
The focus of this chapter is on dosimetric treat- lization in the treatment of a growing list of malig-
ment planning for liver cancer using both of these nancies, there is often a great deal of confusion
radioembolization products, highlighting their among individuals involved in these procedures
similarities and differences. This content will serve on the subject of “dose.” This is at least in part due
as a guide for nuclear medicine physicians, medical to the variable training and backgrounds of the
physicists, and radiation oncologists participating individuals involved in these treatments. The word
and assisting with radioembolization treatment “dose” is a term that is somewhat ambiguous and
planning. is commonly used to describe numerous differing

Table 5.1 SIRTex SIR-Spheres and BTG TheraSpheres: an overview of the properties of 90Y
radioembolization products

SIrtex SIr-Spheres BtG theraSpheres


Composition Resin with Y bound to the
90 Glass permanently
surface impregnated with 90Y
Size (µm) 32.5 ± 5 20–30
Density (g/cc) 1.6 3.3
Number of spheres per vial 40–80 × 106 1.2–8 × 106
activity per sphere at calibration 40–70 Bq 2500 Bq
Long-lived contaminants? No Yes
time postinfusion when 90% of ~9 days ~9 days
absorbed dose is delivered
Shelf-life from calibration 24 hours (Giammarile et al., 15 days (Giammarile et al.,
2011) 2011)
activity per sphere at treatment 50–38.6a Bq 2500–300 Bq (Kritzinger
et al., 2013)
activity per vial at calibration 3 GBq 3, 5, 7, 10, 15, 20 GBq
Source: Salem, R. and Thurston, K. G., J. Vasc. Interv. Radiol., 17, 2006.
a Based on decay at expiration/shelf-life of the product (Giammarile et al., 2011).
5.3 One microsphere or millions of microspheres? / 5.2.3 Effective dose 89

concepts in radioembolization, including absorbed catheter position, and tumor-to-normal uptake


dose, effective dose to the patient or staff, and the ratio (T:N) form the complex relationship relating
dosage delivered to the patient. To prevent confu- dosage to absorbed dose in a particular tissue.
sion, these terms need clear definition in the con-
text of radioembolization. 5.2.3 EFFECTIVE DOSE

5.2.1 ABSORBED DOSE Effective dose is a term that is sometimes used in


the context of radioembolization treatment plan-
The absorbed dose is a physical dose quantity ning. Unfortunately, the term is somewhat confus-
(D) representing the mean energy imparted to ing in the context of radioembolization as the term
matter per unit mass by ionizing radiation. The “effective” does not refer to effectiveness of treat-
SI (International System of Units) unit for the ment for the patient. However, this term is impor-
absorbed dose is the gray (Gy). In radioemboliza- tant for staff members since its primary use is for
tion, as in radiation therapy, the absorbed dose to radiation protection and not for radiation therapy.
the tumor and to uninvolved liver tissues is of pri- The effective dose (E) considers the neoplastic
mary concern. The goal of radioembolization ther- radiation sensitivity of each organ irradiated and
apy is to obtain a sufficiently high absorbed dose combines them into a single term that can be used
to the tumor to produce a therapeutic effect while to estimate cancer risk, that is, the risk of stochas-
limiting the absorbed dose to normal liver tissue, tic effects. For example, effective dose (E) is often
preventing clinically significant toxicity. Of course, used to quantify the increase in cancer risk later in
limiting the absorbed dose to extrahepatic tissues life when a patient receives a diagnostic computed
is also an important concern. This is particularly tomography (CT) scan. It is usually not appropri-
important in the lung and gastrointestinal (GI) ate to apply this term to a patient receiving radia-
tract, where cases of radiation pneumonitis and tion therapy. However, the effective dose can be
GI tract ulceration have been reported (Carretero used to quantify the radiation exposure received
et al., 2007; South et al., 2008a, 2008b; Naymagon by staff members involved in 90Y radioemboliza-
et al., 2010). It is in the calculation of dose to tumor tion procedures, such as nuclear medicine technol-
and nontarget tissue that the term absorbed dose ogists preparing the dose and the interventional
should be used in the setting of radioembolization. radiologists delivering it. This concept is discussed
more in Chapter 7.
5.2.2 DOSAGE
In contrast to “dose” or “absorbed dose,” the term 5.3 ONE MICrOSPHErE
dosage is used to quantify the size or frequency of a
Or MILLIONS OF
dose of a medicine or drug. Ariel’s first radioembo-
lization patients (Ariel and Padula, 1978a, 1978b)
MICrOSPHErES?
were treated with large dosages ranging from 100
to 150 mCi (3.7–5.5 GBq). These dosages were By convention, absorbed dose is used in the set-
larger than those used in modern lobar therapies ting of radioembolization to quantify the average
according to the treatment planning models sug- effects of ionizing radiation within a particular
gested by both major microsphere manufacturers. volume of tissue. In contrast, microdosimetry refers
Because radionuclides are quantified in terms of to the analysis of variable absorbed doses to the tis-
activity, rather than weight or international unit sue immediately adjacent to a single source on a
(IU), treatment dosages of 90Y radioembolization microscopic level. Commonly accepted methods
are quantified in mCi or GBq. The dosage of 90Y calculating average absorbed dose, some of which
radioembolization has a weak correlation to the are discussed later in this chapter, assume uniform
absorbed dose that will be received by the patient’s distribution of microspheres. In reality, the distri-
tumor, normal hepatic parenchyma, or extra- bution of 90Y microspheres is never uniform and the
hepatic tissues. Factors such as liver and tumor absorbed dose varies drastically on a microscopic
volume, infusion technique, lung shunt fraction scale. Microdosimetry sheds light on the complex
(SF), presence of patent gastroenteric collaterals, dose–response relationship in radioembolization
90 Treatment planning part III

and helps to explain major dose–response differ- It is important to keep in mind that radioembo-
ences between radioembolization and external lization therapy involves deposition of millions of
beam radiation therapy (EBRT). individual sources, each capable of widely variable
To illustrate microdosimetry in radioem- dose deposition to the immediately adjacent tissue.
bolization, consider how radiation dose varies The absorbed dose is therefore very heterogeneous
in tissue in the area surrounding a single 90Y when viewed on a microscopic scale. In other
microsphere. Figure 5.1 is a three-dimensional words, there is potential for variable local dose
representation of the dose profile surrounding a deposition at different points within the tumor.
single 90Y microsphere as it decays with analy- However, it is possible to quantify the microscopic
sis performed at the submillimeter scale. Note dose profile from infusion of many microspheres
the extremely heterogeneous nature of the dose if one assumes that 90Y microspheres will deposit
distribution in tissues surrounding this micro- randomly with equal probability in all parts of the
sphere. Nearby tissues and cells will receive large tumor. Figure 5.2 shows a 4 cm mass demonstrated
doses of radiation (>100 Gy) while tissues just a on a magnetic resonance imaging (MRI) with
few millimeters away will receive nonlethal doses. a line subtending the long diameter of the mass.
Figure 5.1 was generated using a Monte Carlo Assuming a randomized, uniform filling pattern
radiation transport code (MCNPX; McKinney of the tumor with 90Y microspheres, the resulting
et al., 2006) with a resolution of 50 µm. Similar absorbed dose profile along the line in Figure 5.2 is
data are available in the literature in the form of shown in Figure 5.3. Note that the dose profile var-
published dose-point kernels (DPK) for varied ies substantially with the number of microspheres
voxel sizes (Strigari et al., 2006; Lanconelli et al., per unit volume in the tumor. Using microspheres
2012) based on calculations using established with a higher specific activity, that is, 2500 Bq
codes such as MCNPX (McKinney et al., 2006) per microsphere versus 50 Bq per microsphere
and FLUKA (Ballarini et al., 2007). (see Table 5.1), will result in a substantially lower

1800
1600
1400
Absorbed dose (Gy)

1200
1000
800
600
400
200
0
5
5
0
0
Distance (mm)
Distance (mm)
5 5

Figure 5.1 Three-dimensional representation of the dose profile surrounding a single 90Y microsphere
as it decays with analysis performed at the submillimeter scale. Note the extremely heterogeneous
nature of the dose distribution in tissues surrounding this microsphere. Nearby tissues and cells will
receive large doses of radiation (hundreds of Gy) while tissues just a few millimeters away will receive
nonlethal doses.
5.4 Distribution in tissue / 5.2.3 Effective dose 91

number of microspheres per unit volume in tis-


sue and, therefore, larger variations in the local
absorbed dose at a microscopic level. The microdo-
simetry of radioembolization is discussed in much
greater detail in Chapter 9.

5.4 DIStrIBUtION IN tISSUE

The simulated absorbed dose profile in Figure 5.3


relies on the assumption that microspheres are ran-
domly deposited in tumor with equal probability, that
is, a microsphere has both the same chance of land-
Figure 5.2 Hepatic protocol MRI demonstrating ing in the center of the tumor and in the periphery.
a 4 cm lesion with a line subtending the short This is rarely true (Fox et al., 1991; Campbell et al.,
diameter of the mass. 2000, 2001; Kennedy et al., 2004). In a pivotal paper

500

450

400

350
Absorbed dose (Gy)

300

250

200

150

100

50,000 spheres/cc at 50 Bq/sphere


50 1,200 spheres/cc at 2500 Bq/sphere
2,500 spheres/cc at 2500 Bq/sphere

0
0 5 10 15 20 25 30 35 40
Distance (mm)

Figure 5.3 Dose profile showing microdosimetric absorbed dose along the profile in Figure 5.2 for
three different average microsphere concentrations. Dose was determined assuming a randomized,
uniform filling pattern of the tumor with 90Y microspheres. Dosimetry was performed with 0.05 mm
resolution.
92 Treatment planning part III

by Campbell et al. (2000, 2001), light microscopy was radioembolization. Large tumor dose heterogene-
used to evaluate the distribution of 90Y microspheres ity ranging between 100 and 3000 Gy was found
directly from microscopic analysis of tissue samples in patients with both primary and metastatic liver
taken from a patient postradioembolization. This cancer treated with glass and resin microspheres,
patient received 3 GBq of 32-µm resin microspheres respectively (Kennedy et al., 2004). Owing to the
to treat an 8-cm metastatic liver tumor. Campbell’s different specific microsphere activity in the glass
analysis noted that the microspheres were highly and resin products, livers treated with resin radio-
concentrated in the periphery of the tumor, creating embolization had more microspheres per cluster
very large absorbed doses ranging from 200 to 600 within the vessels (Kennedy et al., 2004).
Gy. However, the average absorbed dose in the center Evaluation of intratumoral dose heterogene-
of the tumor was only 6.8 Gy. Dose distribution was ity has also been performed noninvasively, using
equally inhomogeneous in uninvolved hepatic tissue various imaging modalities and methods. Three-
with an average dose of 8.9 Gy with approximately dimensional dose distributions can be inferred
1% of normal liver receiving more than 30 Gy. from 99mTc macroaggregated albumin (99mTc-MAA)
The dose heterogeneity determined by micro- single-photon emission computed tomography
scopic analysis of Campbell’s biopsy samples has (SPECT)/CT as also demonstrated by Kennedy
been confirmed on a larger scale with a similar et  al. (2011). In addition, the dose distribution can
analysis of whole livers (Kennedy et al., 2004) after be determined directly from posttreatment 90Y

(a) (b)

(c)

Figure 5.4 (a) Pretreatment contrast-enhanced CT of a patient with cholangiocarcinoma. Left lobe
tumor demonstrates hypodense necrotic core with enhancing peripheral areas of active tumor (b).
Correlating pretreatment 18FDG-PET/CT shows hypermetabolic activity along the peripheral tumor
margin. The necrotic, low attenuating tumor component manifests relatively little FDG avidity. (c) 90Y
PET/CT imaging following embolization of this patient’s left lobe with 821 MBq (22.2 mCi) of resin
microspheres. The low absorbed dose in the center of the necrotic region is plainly visible on post-
treatment 90Y PET/CT.
5.5 Calculating the absorbed dose / 5.2.3 Effective dose 93

bremsstrahlung SPECT or 90Y positron emission 1, high-energy 90Y β-radiation will create second-
tomography/computed tomography (PET/CT), as ary X-rays in the form of bremsstrahlung and char-
described in detail in Chapters 10 and 11. Figure 5.4a acteristic emissions. These photons will penetrate
illustrates the pretreatment contrast-enhanced CT through the liver and contribute to dose in extrahe-
in a patient with cholangiocarcinoma. Figure 5.4b patic tissues and even to individuals in close proxim-
demonstrates the correlating 2-deoxy-2-(18F) fluoro- ity to the patient. However, as discussed by Stabin et
D-glucose (FDG)-PET/CT of the same patient, with al. (1994), this will have little effect on the absorbed
hypermetabolic activity along the peripheral tumor dose in the organ treated with radioembolization. A
margin. The necrotic, low attenuating tumor com- second assumption that becomes important is the
ponent manifests relatively little FDG avidity. Figure permanence of 90Y radioembolization—neither glass
5.4c shows 90Y PET/CT imaging following emboli- nor resin microspheres are biodegradable, and once
zation of this patient’s left lobe with 821 MBq (22.2 infused, both products form a permanent implant.
mCi) of resin microspheres. The low-absorbed dose Combining these two assumptions allows for easy
in the center of the necrotic region is plainly visible calculation of average absorbed dose to an organ of
on posttreatment 90Y PET/CT. Quantification of the interest on a macroscopic scale.
PET data in Figure 5.4c using methods described in This calculation is derived in Equations 5.1
Chapters 11 and 12 yields a maximum tumor dose through 5.3:
of 440 Gy with a minimum dose of just 4.6 Gy at the
necrotic center of the tumor. ∞


Eavg  =  Eϕ ( E ) dE  = 0.935 MeV 
0

= 1.498 ⋅10−13 J (5.1)


5.5 CaLCULatING tHE
aBSOrBED DOSE ∞

Yttrium-90 is one of a handful of radionuclides that


Etot ( J)  =  A0 Eavg ∫e
0
−λt
dt  = 
A0
λ
(
⋅ 1.498 ⋅10−13 )
is considered to be a pure β-emitter, emitting no
gamma rays at any appreciable yield following decay. =  A0 (GBq ) ⋅ 49.86 (J ⋅ s) (5.2)
Although several rare decay pathways of 90Y are dis-
cussed in the following chapters, 90Y is considered a
A0 (GBq ) ⋅ 49.98 (J ⋅ s)
pure β-emitter for all practical purposes related to Davg (Gy )  =     (5.3)
dosimetry. 90Y releases a higher energy electron than M liver (kg)
other pure β-emitters that have been used in inter-
nal radionuclide therapies, with a maximum and where Eavg is the average energy released per decay
average energy of 2.28 and 0.935 MeV, respectively. of 90Y based on the probability density function
The range of the maximum energy 90Y β-particle is (E) for emission (Eckerman et al., 1994), and λ is
11 mm in tissue, while its average energy β particle the Y90 decay constant based on a half-life of 64.24
has a range slightly less than 4 mm. The penetra- hours. A0 is the 90Y activity present in the organ or
tion depth of the high-energy 90Y β-particle is a key organ segment of interest in GBq, and Etot is the total
component of this radionuclide’s success in radio- energy released by A0 from the time that it is infused
embolization, allowing for high dose deposition into until it has fully decayed. The absorbed dose (D) is
the tissues between embolized capillaries. However, expressed in Gy and can be obtained by dividing Etot
when the absorbed dose of 90Y therapy is considered by the mass of the treated liver, Mliver. Alternatively,
for a tumor or nontarget site, an important simpli- the volume of the tissue can be obtained from tomo-
fying assumption can be made: β-radiation released graphic imaging and multiplied by established tissue
from microspheres within a given organ will be fully densities (International Commission on Radiation
absorbed by that organ. This assumption is easy to Units and Measurements [ICRU], 1992). It must
justify based on the average 4-mm 90Y β range in be reiterated that the absorbed dose calculation in
tissue. However, this does not account for second- Equation 5.3 is only valid for 90Y radioembolization
ary radiation, which may extend beyond the 11 mm and only representative of average absorbed dose in
range of a 90Y β-particle. As discussed in Chapter an organ. Equation 5.3 cannot be used for absorbed
94 Treatment planning part III

dose calculation for radioembolization using radio- common following radioembolization (Goin et al.,
isotopes other than 90Y such as 166Ho, which decays 2005; Gulec et al., 2007; Kennedy et al., 2009) and
with the emission of a prompt gamma ray and a does not fit into the common definition of RILD.
β-particle, thus violating the assumption that all Normal liver tissue has a relatively low tolerance
energy emitted during decay is absorbed by the to radiation (Dawson and Guha, 2008). Data from
organ of interest. EBRT suggest that the threshold for RILD follow-
ing whole-liver irradiation is between 30 and 40 Gy
(Emami et al., 1991; Lau et al., 1994; Cremonesi et
al., 2008). Specific cases have demonstrated that liver
5.6 tUMOr aND NOrMaL LIVEr failure can occur from VOD in the setting of EBRT
ENDPOINtS at doses as low as 35 Gy (Sempoux et al., 1997). It is
important to consider that the toxicity thresholds
Careful consideration of tumor and nontarget tissue from EBRT can only be applied to radioembolization
endpoints is paramount to hepatic treatment plan- in a very limited fashion. In fact, the liver can toler-
ning for radioembolization. In addition to nontarget ate higher absorbed doses from radioembolization
hepatic dose deposition, radiation exposure to extra- than it can from fractionated EBRT. This phenom-
hepatic tissues, including the lungs and tissues in the enon may be explained by differences between the
GI tract, are of concern. Consideration of lung dose, dose rate of fractionated EBRT and radioemboliza-
from arteriovenous shunting as well as managing tion, extent of nontumor involved liver, as well as the
patients with extrahepatic nontarget embolization, degree of hypoxia created by capillary occlusion in
is covered in detail elsewhere in this book. As such, radioembolization. In addition, the heterogeneity of
this section will emphasize dosimetry endpoints in absorbed dose at a microscopic scale also contributes
tumor and uninvolved liver tissue. to the decreased hepatic toxicity per gray of radioem-
bolization compared with EBRT due to microscopic
5.6.1 NORMAL LIVER ENDPOINTS sparing of normal tissue. The importance of micro-
scopic dosimetry is discussed in detail in Chapter 9.
Radiation-induced liver disease (RILD) is a term The dose threshold for hepatic toxicity is vari-
that is commonly referenced in the literature as 90Y able among patients and reflects hepatic functional
radioembolization has gained traction as a stan- reserve, liver volume, concomitant diseases and
dard-of-care treatment for primary and metastatic medications, tumor volume, and other patient-
liver cancer. However, the meaning and context of specific factors. The most widely supported upper
RILD are often ambiguous and inconsistent. We absorbed dose toxicity limit to normal liver from
define RILD in patients following radioemboli- radioembolization is 70–80 Gy (Fox et al., 1991;
zation as clinically significant manifestations of Lau et al., 1994; Campbell et al., 2001; Salem and
hepatic insufficiency such as weight gain, ascites, Thurston, 2006). Because underlying cirrhosis
anicteric hepatomegaly, and/or pain in the right decreases the tolerance of the liver to radiation
upper quadrant within 90 days of treatment (Guha (Dawson and Guha, 2008), 70 Gy should be consid-
and Kavanagh, 2011). In patients without under- ered the maximum in cirrhotic patients. However,
lying cirrhosis, these symptoms may be accom- there have been published cases where the absorbed
panied by serologic evidence of hepatic toxicity, dose to normal liver has safely exceeded these
manifested by rising alkaline phosphatase and thresholds, approaching 100 Gy (Gulec et al., 2007).
glutamyl transpeptidase (Sangro et al., 2008). It is Further, treatment planning models for the glass
important to note that RILD represents a spectrum radioembolization product support average doses
of clinical and serologic manifestation of acute of up to 150 Gy in treated tissue (Lewandowski
hepatic injury. RILD is often self-limited, although et al., 2005; Salem and Thurston, 2006).
in its most severe form can result in hepatic vascu- Cirrhosis and other underlying chronic liver
lar endothelial damage leading to a fatal condition disease such as viral hepatitis are not the only con-
resembling veno-occlusive disease (VOD). On the ditions that could decrease the tolerance of the
other end of the spectrum, it should be empha- liver to radiation. It has been shown that previous
sized that Grades 1 and 2 mild liver toxicity is very chemotherapy can increase the occurrence of VOD
5.7 Tumor-to-normal uptake ratio / 5.6.2 Tumor endpoints 95

following radioembolization (Sangro et  al., 2008) trials are currently underway to use advanced
by contributing to intrahepatic vascular endothe- quantitative imaging to identify dose–response
lial damage. Previous research has demonstrated thresholds for metastatic disease. While pub-
that patients with metastatic liver disease receive lished data in the literature is widely varying, it
a higher average dose to nontarget liver even when is likely that neuroendocrine metastases to the
the same dosage of radioembolization is delivered liver or neuroendocrine tumor (NET) are likely
(Sangro et al., 2008). Therefore, one must carefully to show a strong response to radioembolization
consider prior chemotherapy in the treatment at a lower absorbed dose than HCC. On the other
planning process of patients undergoing radio- hand, metastatic colorectal cancer (mCRC) may
embolization for metastatic disease, just as the require an absorbed dose equal to or higher than
severity of cirrhosis affects treatment planning in HCC (Gulec et al., 2007) to achieve the desired
hepatocellular carcinoma (HCC) patients. therapeutic response.

5.6.2 TUMOR ENDPOINTS


5.7 tUMOr-tO-NOrMaL
Tumoricidal endpoints in theory depend on UPtaKE ratIO
many radiobiologic factors, including tumor type,
absorbed dose, heterogeneity of absorbed dose,
and prior radiological or chemotherapeutic treat- The tumor-to-normal uptake ratio (T:N) is an
ments. Because the number of infused micro- important quantity in radioembolization with
spheres and the activity per microsphere also vary glass and resin 90Y microspheres as well as 166Ho
widely between glass and resin microspheres, radioembolization. This quantity has a substan-
dose–response data of one product cannot neces- tial role in treatment planning and can impact the
sarily be applied to the other. tumor-absorbed dose, liver dose, and the toxic-
The largest cohort of published data describing ity or efficacy of the treatment. T:N is defined in
tumoricidal endpoints is for hepatocellular carci- Equation 5.4:
noma. When treating HCC, 120 Gy should be con- A90Y , Tumor / Vtumor
sidered to be a reasonable minimum tumor target T:N =  (5.4)
A90Y , Normal / Vnormal
dose (Yoo et al., 1989; Lau et al., 1994; Ho et al., 1997;
Kennedy et al., 2004; Strigari et al., 2010). Efforts to where A90Y,Tumor is the 90Y activity (MBq) deposited
achieve this absorbed dose in the tumor can be made in the tumor and A90Y,Normal is the activity deposited
using the methods described in the following sec- in uninvolved liver tissue. Vnormal and Vtumor are the
tions, provided the maximum tolerable dose to nor- respective volumes of each. Table 5.2 lists a range
mal liver and extrahepatic tissues is not exceeded. of measured T:N for different tumor types from
Because of the histologic and biologic vari- several publications in the literature.
ability of metastatic liver tumors, dose–response While Table 5.2 shows just a few examples from
data are less certain. However, worldwide clinical the literature, one thing is immediately clear: T:N

Table 5.2 T:N from published sources


t:N Number of
median/ patients or Measurement
Disease mean t:N range tumors method reference
HCC 11.5:1 7:1–16:1 2 Pathologic Kennedy et al. (2004)
HCC 7.0:1 3.9:1–9.2:1 5 99mTc-MAA Gulec et al. (2007)
HCC 3.5:1 1:10–13.5:1 27 99mTc-MAA Lau et al. (1994)
mCrC 6.8:1 2.9:1–15.4:1 15 99mTc-MAA Gulec et al. (2007)
mCrC — 2:5–2:1 2 Pathologic Kennedy et al. (2004)
NEt 5.9:1 3.5:1–11.1:1 20 99mTc-MAA Gulec et al. (2007)
96 Treatment planning part III

varies significantly even in cases of the same as a valid surrogate for 90Y microspheres. There
tumor type. Tumor size, percentage infiltration, are many reasons why this may be untrue, sev-
presence of centralized necrosis, and the tech- eral of which are outlined in Table 5.3.
nique used to measure T:N all contribute to these Many authors have evaluated the validity of
variations. MAA as a radioembolization surrogate, with
no clear consensus (Knesaurek et al., 2010; Kao
5.7.1 USING MAA AS AN et al., 2012; Lam and Smits, 2013; Lam et al., 2013;
ESTIMATION TOOL Wondergem et al., 2013; Garin et al., 2014; Lam
and Sze, 2014). The position of the catheter tip
Before reviewing how T:N can be used in radio- during both infusion of MAA and radioemboliza-
embolization treatment planning, it is impor- tion is among the most critical factors to the prog-
tant to understand methods that can be used to nostic utility of T:N measurements made from
99mTc-MAA SPECT/CT. Positioning of the catheter
quantify it. The most widely used established
method for pretreatment estimation of T:N is tip becomes especially critical when it is near a
MAA SPECT/CT. This technique is convenient bifurcation or when it is positioned in a tortuous
since it is simply an extrapolation of the data vessel (Jiang et al., 2012; Wondergem et al., 2013).
acquired in the pretreatment lung-shunt study. However, in spite of variable correlation between
This method assumes the validity of 99mTc-MAA MAA and 90Y microspheres in the literature, the

Table 5.3 Potential sources of error when using MAA as a microsphere surrogate
Issue Comments Impact amelioration
Specific gravity Significant difference in Moderate N/A
differences between the case of glass
macroaggregated microspheres
albumin (Maa) and
microspheres
Particle size and shape MAA particles generally Moderate N/A
have a wider size range
compared to either
resin or glass
microspheres
Free 99mtc May not significantly Moderate Prophylactic oral
affect T:N administration of 500
measurements but mg perchlorate
could obscure (Ahmadzadehfar et al.,
detection of gastric 2010); use MAA right
nontarget embolization after preparation
(NTE)
Embolic differences Stasis may be reached in High Understand the conditions
some tumors treated under which MAA is
with resin microspheres likely to act as a good
but not MAA surrogate based on
tumor type and size
Catheter position and Catheter positioning High Confirm catheter
centering differences between positioning
MAA and 90Y infusions
can drastically effect
deposition
5.8 Treatment planning strategies / 5.8.1 The empiric model for resin microsphere 97

majority of authors agree that MAA is an excel- from those that allow absolute quantification, such
lent option for treatment planning and predic- as 90Y PET/CT. In 90Y PET/CT, following image
tive dosimetry. Substantial additional discussion acquisition, a region-of-interest drawn in the
related to the use of MAA as a radioembolization tumor will report a value that is directly represen-
surrogate is presented in Chapter 4. tative of the activity of 90Y in that region (Pasciak
et al., 2014). These distinctions are explained in
5.7.2 OTHER TOOLS? detail in Chapter 11.

Cone-beam CT (CBCT) is a promising method


of defining three-dimensional vascular anatomy
and elucidating sources of nontarget embolization 5.8 trEatMENt PLaNNING
(NTE). Louie et al. (2009) evaluated the utility of StratEGIES
CBCT as a tool to augment 99Tc-MAA for the iden-
tification of both extrahepatic NTE and poor T:N. 5.8.1 THE EMPIRIC MODEL FOR
In this study, CBCT was used to identify areas of RESIN MICROSPHERE
extrahepatic localization in 22 out of 42 patients
TREATMENT PLANNING
using CBCT. Perhaps most significantly, CBCT
identified incomplete tumor visualization in 8 out The simplest model for radioembolization treat-
of 42 patients, indicating a secondary vascular sup- ment planning with resin microspheres is the
ply to the tumor (Louie et al., 2009). Cases such as empiric model. The empiric model can be used to
these are often a result of so-called “parasitization” determine treatment dosages for whole-liver radio-
of vascularity, in which the angiogenic factors pro- embolization based purely on the percent hepatic
duced by hypervascular tumors recruit flow from tumor involvement. This model has fallen out of
otherwise insignificant vessels. This phenomenon is favor and its clinical use has been largely replaced
common following one or more chemoembolization by preferred methods such as the body surface area
treatments, which preferentially depend on small (BSA) methods and partition models, discussed
vessel embolization to achieve treatment efficacy. If in the following sections. However, it should be
identified during pretreatment angiography, collat- noted that the empiric model is still included on
eral vessels may be prophylactically embolized, thus the SIRTeX SIR-Sphere package insert (SIRTeX
redirecting more vascular flow to the treated vessel Technology Pty, Lane Cove, NSW, Australia). In
and increasing the likelihood of technical success. addition, the empiric model was the treatment
In extreme cases, extrahepatic collateral vessels strategy used during the initial SIRTeX SIR-Sphere
may provide the majority of a tumor’s blood supply, clinical trials.
and standard delivery of 90Y microspheres via the In the largest single cohort of retrospective data
hepatic arteries can result in a T:N less than 1. In of patients who underwent radioembolization using
situations such as these, CBCT can be a powerful resin microspheres, 28 expired with a cause of death
tool in treatment planning and delivery. attributable to complications related to liver toxicity.
CBCT has also been investigated as a poten- Of these deaths, 21 were from a single center that
tial tool in quantifying T:N. A study by Jones and used the empiric model exclusively for treatment
Mahvash (2012) used gelatin phantoms injected planning (Kennedy et al., 2009). This does not sug-
with different concentrations of iodinated contrast gest that the empiric model is unsafe; however; it is
to show that CBCT is able to quantify relative con- imperative that the model be used correctly.
trast enhancement. Since T:N is a ratio, absolute When using the empiric model, it is critical
quantification is not necessary for its calculation. to understand that its definition in Table 5.4 is
In other words, relative densities in tumor and for whole-liver radioembolization. For lobar or
normal liver in pre- and postcontrast CBCT can segmental therapy, one must modify the recom-
be effectively used to estimate T:N. In this man- mended dosage (Aemp) in Table 5.4 according to
ner, CBCT can be used as an excellent way to aug- Equation 5.5:
ment 99mTc-MAA scintigraphy in the estimation of
 V 
T:N for use in partition model treatment planning. A ( GBq ) =   Aemp ( GBq ) ⋅  treated  (5.5)
One must be careful to differentiate this method V
 wholeliver 
98 Treatment planning part III

Table 5.4 Description of the empiric model used patients treated with resin microspheres (Kennedy
for 90Y radioembolization with resin et al., 2009).
microspheres When using the BSA model, it is critical to
understand that its definition in Equations 5.6 and
the percentage of tumor recommended 5.7 is for whole-liver radioembolization. For lobar
involvement in the liver dosage, Aemp (GBq) or segmental therapy, one must modify the recom-
More than 50% 3.0 GBq mended dosage, ABSA, in Equation 5.7 according to
25%–50% 2.5 GBq Equation 5.8:
Less than 25% 2.0 GBq
 V 
A ( GBq ) =   ABSA ( GBq ) ⋅  treated  (5.8)
 Vwholeliver 
where Aemp is the recommended dosage from the
empiric model as defined in Table 5.4. Vtreated and where ABSA is the recommended dosage from the
Vwhole liver are the volumes of the portion of the liver BSA model as defined in Equations 5.6 and 5.7.
to be treated and entire liver volume, respectively. Vtreated and Vwhole liver are the volumes of the portion
of the liver to be treated and entire liver volume,
respectively.
5.8.2 THE BSA MODEL FOR RESIN
MICROSPHERE TREATMENT
PLANNING 5.8.3 GLASS MICROSPHERE
TREATMENT PLANNING
The most widely used treatment planning tech-
nique for radioembolization with resin micro- Standard treatment planning for radioemboli-
spheres is the BSA method. This method may have zation using glass microspheres is a relatively
some familiarity to many clinicians since BSA- straightforward process, giving treating physi-
based calculations are widely used in medicine for cians significant latitude to consider patient-
determining dosages for medications such as che- specific factors. The foundational principle is
motherapy. Treatment dosage using the BSA model based on Equation 5.3, which describes the
is strongly dependent on the patient’s height and average dose in a tissue volume as a function
weight and moderately dependent on the percent of 90Y dosage. Equation 5.3 can be rewritten to
tumor infiltration. Equations 5.6 and 5.7 describe solve for the treatment dosage, Ao, as shown in
the BSA treatment planning method: Equation 5.9:

BSA (m 2 ) =  0.2025 ⋅ height 0.725 ( m )


Davg (Gy ) ⋅ M liver (kg)
Ao (GBq )  =      (5.9)
⋅ weight 0.425 (kg) 49.98(J ⋅ s)
(5.6)
where Davg is the target-absorbed dose in the por-
 Vtumor  tion of the liver to be treated and Mliver is the mass of
ABSA  (GBq ) = BSA − 0.2 +   (5.7)
 Vtumor + Vnormal  treated liver tissue. Liver mass is extrapolated from
volumetric analysis performed on pretreatment
Vtumor and Vnormal are the respective volumes of CT data for planning lobar therapy. In the setting
tumor and uninvolved liver tissue in the portion of radiation segmentectomy, segmental volume
of the liver to be treated. The BSA model assumes may be calculated by preprocedural cross-sectional
a relationship between the physical size of the imaging or from CBCT during the pretreatment
patient and ability to tolerate increasing dosage. mapping procedure, discussed in additional detail
The concept that larger patients (not necessarily in Chapter 6. Once hepatic volume is obtained, a
with larger livers) are more tolerant to increased conversion factor of 1.05 kg/L is used to convert
dosages of 90Y has been shown in the literature hepatic tissue volume to mass (ICRU, 1992).
(Sangro et al., 2008). The BSA model was also The average dose endpoint (Davg) used in treat-
found to have a lower risk of liver toxicity than the ment planning is recommended to be in the
empiric model in the aforementioned cohort of 680 range between 80 and 150 Gy as specified in the
5.8 Treatment planning strategies / 5.8.5 Comparing treatment planning models 99

Therasphere package insert (BTG International A0 (GBq ) ⋅ 49.98 ⋅ FU tumor


Ltd., London, UK). The endpoint used should be Dtumor (Gy ) =    (5.13)
M tumor (kg)
selected based on tumor burden, health of unin-
volved liver tissue, previous therapy, and other
clinical factors that may affect toxicity and/or where Ao is the activity of 90Y to be infused into
response. the liver. Thus, Equation 5.13 can be rearranged to
Finally, it should be mentioned that Equation derive the prescribed 90Y dosage in Equation 5.14:
5.9 does not account for lung shunting or residual
activity remaining in the delivery system, both of   Dtumor (Gy ) ⋅ M tumor ( kg )
A0 (GBq ) =    (5.14)
which will decrease the dosage of radioactivity 49.98 ⋅ FU tumor
deposited in the liver, Ao, and therefore Davg.
The partition model equation uses patient-
specific tumor and liver volumes, along with pre-
5.8.4 THE PARTITION MODEL determined T:N from pretreatment 99mTc-MAA
SPECT/CT and/or CBCT. Thus, this method
The partition model for radioembolization (Ho represents the most tailored treatment planning
et al., 1996, 1997), also referred to as the medi- algorithm, allowing for accurate estimation of
cal internal radiation dose (MIRD) model (Gulec absorbed dose to tumor, nontarget liver tissue, and
et al., 2006), is a three-compartment model that lungs. Prior research has shown that treatment
takes into account patient-specific data to tailor planning with the partition model based on 99mTc-
the desired dosimetric endpoints in the tumor, MAA SPECT/CT can improve clinical outcomes
normal liver, and lungs. Unlike the previously (Gulec et al., 2006).
discussed models that are specific to resin or glass As discussed in a previous section, when treating
microspheres, the partition model can poten- an HCC patient with radioembolization, one may
tially be used for either product. However, one wish to set Dtumor to a minimum of 120 Gy. Solving
must always keep in mind that tumor and nor- for Ao, however, is not the only necessary step in
mal liver dose–response relationships are likely the treatment plan. A0 must be back-substituted
to vary between resin and glass products due to into Equation 5.11 to ensure that the absorbed dose
variable-specific activity, sphere number/density, to normal liver tissue is below suggested limits.
and sphere composition. In addition, the dosimetric techniques described
The lung SF is an important input into the parti- in chapter 4 should also be applied to ensure an
tion model that is used along with T:N, tumor vol- acceptable lung dose.
ume (Vtumor), and nontarget liver volume (Vnormal) to It is critical to note that the partition model can be
determine the fractional uptake (FU) into a com- effectively used for any radioembolization product
partment. Equations 5.10 and 5.11 are the equa- including resin or glass 90Y microspheres and 166Ho
tions for the calculation of FU in the uninvolved radioembolization. However, tumor and uninvolved
liver and tumor: liver efficacy and safety endpoints are likely to differ
between radioembolization products.
 Vnormal 
FU normal = (1 − SF)   (5.10)
 T:N ⋅ Vtumor +  Vnormal  5.8.5 COMPARING TREATMENT
PLANNING MODELS
 T:N ⋅ Vtumor 
FU tumor = (1 − SF)   (5.11) In the previous sections, four treatment planning
 T:N ⋅ Vtumor +  Vnormal  models have been introduced for radioembolization
using resin microspheres, glass microspheres, or
The FU can be used to determine the average
both. However, it is difficult to understand simply by
absorbed dose in both tumor and normal liver com-
examining the equations how the treatment activ-
partments according to Equations 5.12 and 5.13:
ity prescribed using these models differs in patients
of varying size, tumor burden, and T:N. Figure 5.5a
A0 (GBq) ⋅ 49.98 ⋅ FU normal
Dnormal (Gy) =    (5.12) compares the recommended dosage of resin micro-
M normal (kg) spheres based on the empiric, BSA, and partition
100 Treatment planning part III

4 4

3.5 3.5

3 3

2.5 2.5
Dosage (GBq)

Dosage (GBq)
2 2

1.5 1.5

1 1
SIRSphere empirical model
th
SIRSphere BSA model, female in 10 percentile of height/weight
th
SIRSphere BSA model, female in 50 percentile of height/weight Therasphere 80 Gy treatment endpoint
0.5 th
SIRSphere BSA model, male in 50 percentile of height/weight 0.5 Therasphere 120 Gy treatment endpoint
th Therasphere 150 Gy treatment endpoint
SIRSphere BSA model, male in 90 percentile of height/weight
Partition model, 2:1 T:N, 150 Gy tumor dose Partition model, 2:1 T:N, 150 Gy tumor dose
Partition model, 5:1 T:N, 150 Gy tumor dose Partition model, 5:1 T:N, 150 Gy tumor dose
0 0
0 10 20 30 40 50 60 70 0 10 20 30 40 50 60 70
Percentage tumor burden (%) Percentage tumor burden (%)
(a) (b)

Figure 5.5 Prescribed dosage recommendation for (a) resin microspheres and (b) glass microspheres
as a function of patient size, tumor burden, and T:N. Data are presented for a 1300 mL liver. Patient
size data are from published U.S. census demographic information 2007–2008.

models as a function of patient size, tumor burden, than the other since toxicity and efficacy endpoints
and T:N. One can immediately see that the empiric differ between the two products. Chapter 9 will
model yields a dosage that is higher than most of explain this phenomenon at a microscopic level.
the other models, and it does this regardless of cir-
cumstance (i.e., varying patient size or liver size).
On the other hand,  patient size (height  +  weight) 5.9 CONtOUrING LIVEr aND
has a large impact on the BSA treatment planning tUMOr VOLUMES
model. A female in the 10th percentile of height
and weight will be treated with about half the dos- Each of the treatment planning methodolo-
age as a male in the 90th percentile of height and gies presented in the previous section require
weight. The partition model with a T:N of 5:1 yields measurement of tissue volume through some form
the lowest dosage at a low percentage tumor infiltra- of volumetry based on CT, MR, or hybrid (e.g., PET/
tion but rises steeply as the tumor burden increases. CT) tomographic imaging. The empiric and BSA
Use of the partition model with a low T:N (2:1 or models are reliant on measurement of the percent
less) can result in a large dosage and the potential tumor infiltration and also the percentage of treated
for high absorbed doses to uninvolved liver. Figure liver tissue relative to total liver volume. Treatment
5.5b describes the dosage recommended for glass planning for glass microspheres depends strongly
treatment planning at 80, 120, and 150 Gy absorbed- on the total volume of liver tissue being treated,
dose endpoints. Comparing Figure 5.5a with 5.5b, while the partition model relies on both volume of
it is quickly apparent that larger dosages are used tumor and uninvolved liver tissue. These volumes
for radioembolization with glass microspheres than can be determined using automatic, semiautomatic,
resin microspheres. However, this does not suggest or manual techniques as described in Sections 5.9.1
that one product carries less toxicity or more efficacy and 5.9.2.
5.9 Contouring liver and tumor volumes / 5.9.2 Manual segmentation 101

5.9.1 AUTOMATIC AND automation of atlas-based segmentation is greater, in


SEMIAUTOMATIC part, because it is reliant on a standard atlas of refer-
SEGMENTATION ence segmented organ contours. These contours are
predefined and validated by radiologists or anato-
Automatic and semiautomatic organ and tumor mists. The atlas is then matched to the dataset of
segmentation have been widely used in areas of interest using deformable registration. To improve
health physics, radiation therapy, radiology, surgery, the accuracy of the process and account for variant
and other facets of medicine for years with different anatomy, multiatlas-based segmentation is often
levels of accuracy and required human input (van employed. In this technique, many deformable regis-
Ginneken and Haar Romeny, 2000; Ghanei et al., trations are performed with atlases that feature vary-
2001; Marroquín et al., 2002; Caon, 2004; François ing anatomy and those producing the best match
et al., 2004; Buie et al., 2007; Fripp et al., 2007; Klein are combined or fused together to produce a final
et al., 2008; Metzger et al., 2013; Kockelkorn et al., optimal segmentation. Several fusion methods are
2014). Techniques such as thresholding and region employed with the simplest a majority vote fusion to
growing (El-Baz et al., 2011) can be quite effective more complex techniques that incorporate statistical
in high-contrast anatomical areas that are clearly models into the fusion process (Warfield et al., 2004).
demarcated. These techniques have even been used In the context of radioembolization treatment
successfully in the semiautomatic segmentation of planning, automated delineation of lobar or seg-
the liver based on CT imaging (Foruzan et al., 2009). mental liver volumes can significantly speed the
In the context of radioembolization treatment planning process. While atlas-based segmentation
planning, available segmentation tools will dictate usually requires some postsegmentation manual
the degree of automation that can be achieved in the correction (Figure 5.6), this task can be accom-
clinical workflow. However, some automation can be plished much faster than manual segmentation
achieved in almost every clinical environment with- alone. Small inaccuracies in the measured liver lobe
out specialized tools. For example, consider patients volume, segment volume, or tumor volume should
with metastatic liver disease who have received a be considered in the context of the sensitivity of the
pretreatment 18FDG-PET/CT exam. In many of treatment planning model to volume changes dis-
these patients, manually contouring multiple liver cussed in Section 5.8.5 (Figure 5.5).
lesions to determine the percent tumor involvement
can be a time-consuming task. However, determin-
ing the tumor burden is a necessary component of 5.9.2 MANUAL SEGMENTATION
treatment planning for radioembolization using
resin microspheres. Automated 3D thresholding At institutions where radioembolization treatment
tools available on basic radiology reading stations planning is not assisted by a radiation oncology
can be used in conjunction with 18FDG-PET/CT to department, sophisticated thresholding and segmen-
delineate hypermetabolic tumor boundaries. The tation tools may be unavailable. In these cases, man-
threshold can be adjusted on a per-patient basis or ual segmentation may be the only option. The authors
set quantitatively as a function of maximum stan- of this chapter have found that DICOM viewers that
dardized uptake value (SUVmax). This technique feature spline-based regions of interest (ROIs) com-
is useful for many patients with FDG-avid meta- bined with interpolative volumetry can significantly
static liver tumors treated with radioembolization. reduce the time required for manual liver and tumor
An example of thresholding at 40% of whole-liver delineation since ROIs need not be drawn on every
SUVmax is shown in Figure 5.8b. slice. The three-dimensional liver volume rendered
If automated segmentation tools are avail- in Figure 5.7 was generated with contours on only six
able, atlas-based segmentation (Ghanei et al., 2001; axial CT slices using the free DICOM viewer OsiriX
Bondiau et al., 2005; Zhang et al., 2006; Reed et 5.8.2 (Osirix Foundation, Geneva, Switzerland).
al., 2009; Linguraru et al., 2010; El-Baz et al., 2011; Once again, errors in either tumor or lobe delin-
Park et al., 2014) is a method that has the potential eation resulting from manual contouring with or
for more accurate organ segmentation and may without interslice interpolation should be weighed
require less user input compared to alternatives such against the sensitivity of the treatment planning
as thresholding and region growing. The degree of model as discussed in a Section 5.8.5 (Figure 5.5).
102 Treatment planning part III

(a) (b)

Figure 5.6 (a) Results of fully automated atlas-based segmentation of the left and right liver lobes
using the MIM 6 (MIM Software Inc., Cleveland, OH) software package. (b) Atlas-based segmentation
followed by manual correction (shaded areas).

Figure 5.7 Three-dimensional liver volume generated with contours on only six axial CT slices using
the free DICOM viewer OsiriX 5.8.2 (Osirix Foundation, Geneva, Switzerland).
5.10 Treatment planning calculations / 5.10.1 Lobar therapy 103

on  the portal venous phase CT and manual


5.10 trEatMENt PLaNNING segmentation of the CT image set would be lim-
CaLCULatIONS ited to corresponding hypodensity visualized in
Figure 5.8a. The 18FDG-PET/CT identifies the
This section functions as a guide that will pro- tumor as centrally necrotic and highly FDG-
vide instructive examples of radioembolization avid along its periphery. Due to these differences,
treatment planning in both lobar and segmental manual contouring from contrast-enhanced CT
therapies. Different treatment planning models are and semiautomated threshold-based contouring
considered for both resin and glass microspheres. based on PET/CT yielded left lobe tumor vol-
umes of 67 and 122 mL, respectively. These vol-
umes include out-of-slice lesions not visualized
5.10.1 LOBAR THERAPY
in Figure 5.8.
As a surgeon aims to resect a margin of normal
parenchyma surrounding a liver tumor, radioembo- 5.10.1.2 Scenario 1: Empiric model
lization is often purposefully nonselective, so as to for resin microspheres
treat microscopic metastatic disease. Patients with
liver metastases are usually noncirrhotic and have a Based on these measurements, the percent-
greater functional hepatic reserve, and are therefore age tumor infiltration in the liver is as follows in
commonly treated with microsphere infusion to an Equations 5.15 and 5.16 for the tumor volumetry
entire hepatic lobe. Lobar therapy is also desirable in performed on CT and PET/CT, respectively:
the setting of liver metastases due to the propensity
for multifocal disease, making superselective infu- Vtumor 67 mL
I tumor,CT = =   ×  100%
sion impractical. It should be noted that this meth- Vtumor + Vnormal 424 mL
odology applies to HCC patients with multifocal = 15.8% (5.15)
disease in the absence of clinical and serologic indi-
Vtumor 122 mL
cators of severe underlying liver disease. However, I tumor,PET = = ×  100%
in the setting of underlying liver impairment, mini- Vtumor + Vnormal 424 mL
mizing nontarget embolization to nontumor hepatic = 28.7% (5.16)
tissue is preferred, so as to preserve hepatic function
(Salem and Thurston, 2006). It is important to note in Equations 5.15 and
5.16 that the percent infiltration was calculated
only for the lobe to be treated, that is, using the vol-
5.10.1.1 Example case ume of tumor and volume of normal liver within
A 61-year-old female presents with lung cancer the left lobe only. As denoted in Table 5.4, the rec-
and four metastatic lesions to the left hepatic lobe. ommended whole-liver treatment dosage (Aemp)
There is no serologic or radiologic evidence of cir- for this level of infiltration is 2.0 and 2.5 GBq, for
rhosis or biliary obstruction. Figure 5.8a shows a 15.8% and 28.7% tumor infiltration, respectively.
hepatic protocol CT of one centrally necrotic met- Recall that the empiric model is defined for whole-
astatic lesion in the left hepatic lobe, with corre- liver therapy and the dosage must be scaled for the
sponding hypermetabolic activity by 18FDG-PET/ treated lobe. Equation 5.17 shows this calculation
CT (Figure 5.8b). In all treatment planning sce- for the 2.5 GBq prescription:
narios (1–4 below) a left lobar therapy is assumed
using conventional anatomic  boundaries. Each  V 
A ( GBq ) =   Aemp ( GBq ) ⋅  treated  = 2.5
lobe was contoured in three dimensions to deter-  Vwholeliver 
mine relative volumes  of the left lobe, right lobe,
424 mL
GBq ⋅ 
and total liver (Figure 5.8c). =
 0.75 GBq
The volume of the left lobe tumors can be  986 mL  +  424 mL  (5.17)
contoured based on the contrast-enhanced CT or
18FDG-PET/CT (Figure 5.8a and b). In this par- The final step illustrated in Equation 5.17 is crit-
ticular case, the tumor is not avidly enhancing ical. This can be appreciated using Equation 5.3 to
104 Treatment planning part III

(a) (b)

(c) (d)

Figure 5.8 (a) Hepatic protocol CT performed on a patient with lung cancer metastases to the liver.
A centrally necrotic metastatic lesion is present in the lateral left hepatic lobe (a) and correspond-
ing hypermetabolic activity by 18FDG-PET/CT is shown in (b). Automatic contouring of the lesion was
performed in (b) based on a threshold of 40% of SUVmax (blue line). (c) The hepatic protocol CT was
used to contour both the left and right lobe volume (white line) as well as the tumor based (blue line)
on anatomical boundaries. (d) Pretreatment 99mTc-MAA SPECT with tumor boundary (blue line) and
normal left lobe tissue region (white line).

calculate the average absorbed dose to the left liver 1.62 m (5 feet, 4 inches). Therefore, her body surface
lobe if the step in Equation 5.17 is not performed. area can be calculated according to Equation 5.6:
Applying a density of 1.05 kg/L to this patients’
424 mL left lobe, we obtain a mass of 0.445 kg. BSA (m 2 ) =  0.2025 × height 0.725 (m) ⋅
A dosage of 2.5 GBq, according to Equation 5.3, weight 0.425 (kg)
would yield an average absorbed dose of 280 Gy to
= 0.2025 × 1.620.725 (m) ⋅ 640.425 (kg)
the lobe. This level of absorbed dose would likely
result in severe toxicity in the uninvolved tissues of =   1.68 m 2   (5.18)
that lobe.
One must keep in mind that the units of height
(m) and weight (kg) are critical as shown in
5.10.1.3 Scenario 2: Resin Equation 5.18:
microspheres using the  Vtumor 
BSA model Absa  ( GBq ) =  BSA − 0.2 +    
V
 tumor + Vnormal 

Now let us change gears and consider the BSA treat- 122 mL
=  1.68  –  0.2  +  
ment planning model for this scenario. This female 424 mL
patient has a weight of 64 kg (141 lb.) and a height of = 1.767 GBq (5.19)
5.10 Treatment planning calculations / 5.10.1 Lobar therapy 105

In Equation 5.19, the BSA dosage (ABSA) has been disease or biliary obstruction, any endpoint in
calculated based on the tumor volumes identified 100–120 Gy would likely be appropriate.
using 18FDG-PET/CT. ABSA would be slightly lower
had the tumor volumes from contrast-enhanced
CT been used. 5.10.1.5 Scenario 4: Resin
Once again, the BSA model is defined for microspheres using the
whole-liver therapy and the dosage (ABSA) must be partition model
scaled for the treated lobe. This process is shown
in Equation 5.20. Note that the final recommended Lobar therapies will result in microsphere deposi-
treatment activity using the BSA method (0.53 tion in normal liver parenchyma and microsphere
GBq) is less than when the empiric method is used deposition in the tumor according to the T:N
(0.75 GBq): ratio. The partition model accounts for measured
T:N as well as normal liver and tumor volumes to
 V  design a patient-specific treatment plan. Figure
A ( GBq ) =   ABSA ( GBq ) ⋅  treated  5.8d describes the measurement process of T:N
V
 wholeliver  following 99mTc-MAA SPECT/CT imaging. A left
 424 mL  lobar infusion of 150 MBq of 99mTc-MAA was per-
= 1.767 GBq ⋅  
986 mL  +  424 mL formed 2 weeks before treatment with the catheter
 
tip positioned carefully at the expected treatment
= 0.53 GBq (5.20)
location. Tumor contours from the metabolically
active region on 18FDG-PET/CT (Figure 5.8b) have
been copied onto the 99mTc-MAA SPECT image set
5.10.1.4 Scenario 3: Glass (Figure 5.8d). Areas of uninvolved liver are also
microspheres using the shown. T:N was calculated in this case according
to Equation 5.4 to be 2.8:1. Based on the prether-
recommended treatment
apy MAA mapping procedure, a lung SF for this
planning method patient was measured to be 5%. With this infor-
mation, we can calculate the FU in tumor and
Treatment planning for the left lobe therapy of the
uninvolved liver.
same patient using glass microspheres is consid-
To compute the FU, we must determine the
erably more straightforward. The percent tumor
volume of uninvolved parenchyma in the left lobe
burden is not necessary and the solution is depen-
(Vnormal), which was not previously included in
dent only on the total mass of tissue in the left lobe:
Table 5.5. If we assume tumor volumes based on
0.445 kg. In fact, the most difficult part is select- 18FDG-PET/CT, then V
normal = 424–122 mL = 302
ing an absorbed dose endpoint within the recom-
mL. The corresponding mass of uninvolved left
mended 80–150 Gy range.
lobe parenchyma is 0.317 kg:
For the purposes of this example, selecting a
conservative 100 Gy absorbed dose treatment end-  
Vnormal
point, we obtain FU normal = (1 − SF )  
 T:N ⋅ Vtumor +  Vnormal 

Davg (Gy ) ⋅ M liver (kg)  302 mL 


A0 (GBq )  =   = (1 − 0.05 )  
49.98 (J ⋅ s)  2.8 ⋅ 122 mL + 302 mL 
100 Gy ⋅ 0.445 kg = 0.45  (5.22)

49.98 T
 
= 0.89 GBq  (5.21)  ⋅ Vtumor 
FU tumor = (1 − SF )  N
T:N ⋅ Vtumor +  Vnormal 
 
Note that the absorbed dose endpoint shall  
be selected based on the many clinical and sero-  2.8 ⋅ 122 mL 
=  (1 − 0.05 ) 
logic factors discussed in the case introduction.  2.8 ⋅ 122 mL + 302 mL 
However, in this patient without chronic liver = 0.50 (5.23)
106 Treatment planning part III

Table 5.5 Volumetric measurements of the patient presented in Figure 5.8


Structure Volume (mL) Mass (kg)a
Total liver 1,410 1.480
Total right lobeb 986 1.035
Total left lobeb 424 0.445
Left lobe tumor, manually contoured 67 0.070
from CT
Left lobe tumor, automatically measured 122 0.128
from 18FDG-PET/CT with threshold =
0.4 · SUVmax
Right lobe tumor 0 mL 0 kg
Note: The patient has a weight of 64 kg (141 lb.) and a height of 1.62 m (5 feet, 4 inches).
a Mass calculated by multiplying volume (L) by the density of liver tissue, 1.05 (kg/L) (ICRU, Photon, Electron,

Proton and Neutron Interaction Data for Body Tissues, International Commission on Radiation Units and
Measurements, 1992).
b Includes contributions from uninvolved tissue and tumor tissue.

We can now determine the activity required to therapy due to the small fraction of liver tissue irra-
reach a tumor-absorbed dose endpoint as shown diated. This particular point is carefully detailed in
in Equation 5.24. Given that this patient has met- Chapter 8.
astatic liver cancer with no underlying chronic
liver disease, a tumor-absorbed dose of 150 Gy 5.10.1.6 Other considerations
for radioembolization using resin microspheres is
appropriate: Each case above represented an example of treat-
ment planning for a single-session therapy.
  Dtumor (Gy ) ⋅ M tumor ( kg ) Fractionation of radioembolization treatments is a
A0 (GBq ) =  recommended practice, which may decrease tox-
49.98 ⋅ FU tumor
icity to normal liver (Salem and Thurston, 2006;
150 (Gy ) ⋅ 0.128 (kg)
=  Cremonesi et al., 2008). Multicycle therapy will be
49.98 ⋅ 0.50 discussed in more detail in Chapter 8.
= 0.77 GBq (5.24)

Assuming accuracy of T:N measurement, a 5.10.2 SELECTIVE THERAPY


dosage of 0.77 GBq will result in a 150 Gy aver-
Segmental and selective therapies will result in
age tumor dose. However, this dosage must still
microsphere deposition in tumor and normal tis-
be back substituted into Equation 5.12 to ensure
sues within the segment according to T:N and
that normal liver toxicity thresholds are not
should yield little or no deposition outside of that
exceeded. This is illustrated in
segment except in cases of reflux. While outside
A0 ( GBq ) ⋅ 49.98 ⋅ FU normal the scope of this chapter, vascular planning for
Dnormal ( Gy ) =   segmental therapy has been discussed elsewhere
M normal (kg)
(Salem and Thurston, 2006; Riaz et al., 2011) and
0.77 ( GBq ) ⋅ 49.98 ⋅ 0.45 in Chapter 3. Instead, this section will focus pri-

0.317 (kg) marily on dosimetric aspects of selective therapy.
= 55 Gy  (5.25) In all cases of lobar therapy reviewed in the previ-
ous section, treatment planning was always depen-
As previously discussed, the maximum tol- dent on the volume of the treated lobe. In selective
erable dose to normal, noncirrhotic, liver tissue and segmental therapy, one must instead identify
from radioembolization is approximately 80 Gy. A the volume of tissue perfused by the treatment that
treatment dosage of 0.77 GBq in this case is likely may consist of only tumor or tumor and normal
to be safe, particularly in the setting of a left-lobe liver tissue within one or more segments.
5.10 Treatment planning calculations / 5.10.2 Selective therapy 107

As shown in Figure 5.9, the patient has a 22 cystic carcinoma (ACC). The patient’s total liver
cm solitary liver metastasis from parotid adenoid volume is 3500 mL, with 2200 mL attribut-
able to tumor. Contrast-enhanced CT and pre-
treatment angiography suggest that the tumor
is well perfused and solid, with no substantial
areas of necrosis. This patient was referred for
radioembolization using resin microspheres.
Unfortunately, this case represents a clinical and
technical challenge owing to the large tumor size.
Targeting of this patient’s entire tumor would
result in a subtumoricidal response, even if the
entire 3 GBq resin microsphere dosage was deliv-
ered. Therefore, the tumor was treated in mul-
tiple stages. Figure 5.10a shows the pretreatment
angiography with the catheter positioned in the
right hepatic artery and two dominant branches
supplying the tumor. Figure 5.10b shows a selec-
tive segmental right hepatic arteriogram prior to
99mTc-MAA injection. The 99mTc-MAA SPECT/

CT in Figure 5.11 shows the volume of tumor


Figure 5.9 Contrast-enhanced CT of a patient perfused to be 48% of the total tumor volume.
with a 22 cm solitary liver metastasis from parotid Since MAA deposited almost exclusively in the
ACC. Contrast-enhanced CT and pretreatment
tumor, Equation 5.3 could be used directly to
angiography suggest that the tumor is well
determine the dosage necessary to reach a 120 Gy
perfused and solid, with no substantial areas of
necrosis. endpoint in 48% of the tumor volume:

(a) (b)

Figure 5.10 (a) Pretreatment angiography with the catheter positioned in the right hepatic artery and
two dominant branches supplying the tumor. (b) Selective segmental right hepatic arteriogram prior
to 99mTc-MAA injection.
108 Treatment planning part III

  Dtumor (Gy ) ⋅ M tumor ( kg )


A0 (GBq ) = 
49.98
120 (Gy ) ⋅ 0.48 ⋅ 2.2 (L ) ⋅1.05 (kg / L)

49.98
= 2.66 GBq (5.26)

Large tumor size is certainly not the only rea-


son to treat in stages. A segmental approach to the
treatment of multifocal disease is also an excellent
option, which can reduce toxicity in normal liver
(Riaz et al., 2011). Figures 5.12a and 5.12b show a
hepatic protocol MRI and 111In Octreotide SPECT
of a patient with NET metastases. Multiple meta-
static lesions are present throughout the right lobe.
Figure 5.11 99mTc-MAA SPECT/CT showing the
Given the absence of underlying chronic liver dis-
volume of tumor perfused at the catheter loca- ease and the typically high efficacy of radioem-
tion shown in Figure 5.10b. Perfused volume of bolization in treating NET, a selective multistep
MAA was measured to be 48% of the total tumor approach was employed. A lateral right hepatic
volume. artery branch was selected (Figure 5.12c and d)

(a) (b)

(c) (d)

Figure 5.12 (a) Hepatic protocol MRI and (b) 111In octreotide SPECT performed on a patient with
neuroendocrine metastases to the liver. Multiple metastatic lesions are present throughout the right
lobe. (c) Angiogram showing right hepatic arterial vasculature. (d) Selective microcatheter positioning
in lateral right hepatic arterial branch that, based on pretreatment 99mTc-MAA SPECT, perfused the
posterior half of the right lobe volume.
References 109

a  team-based approach to treatment planning. A


periodic meeting to discuss upcoming cases con-
sisting of the treating interventional radiologist,
medical physicist, nuclear medicine physician,
and/or radiation oncologist will help to elucidate
optimal treatment plans through multidisciplinary
discussion. While such meetings can be difficult to
schedule in a busy clinical setting, their importance
cannot be overemphasized and will undoubtedly
lead to improved patient outcomes.

REFERENCES
Ahmadzadehfar, H., Sabet, A., Biermann, K.
et al. (2010). The significance of 99mTc-MAA
Figure 5.13 Posttreatment 90Y PET/CT verifying
SPECT/CT liver perfusion imaging in treatment
that the radioembolization dosage was delivered
planning for 90Y-microsphere selective internal
to the posterior half of the right hepatic lobe.
radiation treatment. J Nucl Med 51:1206–1212.
Ariel, I.M., Padula, G. (1978a). Treatment of
that, based on pretreatment 99mTc-MAA SPECT, symptomatic metastatic cancer to the liver
perfused approximately 50% of the right lobe vol- from primary colon and rectal cancer by the
ume. Treatment planning was performed accord- intra-arterial administration of chemotherapy
ing to the BSA model for radioembolization using and radioactive isotopes. Prog Clin Cancer
resin microspheres. The dosage recommended by 7:247–254.
the BSA model was reduced by 50% due to the esti- Ariel, I.M., Padula, G. (1978b). Treatment of symp-
mated perfused volume of the treatment territory. tomatic metastatic cancer to the liver from
Following delivery of 0.81 GBq of 90Y, a 90Y PET/ primary colon and rectal cancer by the intra-
CT scan was performed verifying limited micro- arterial administration of chemotherapy and
sphere deposition outside of the treatment zone radioactive isotopes. J Surg Oncol 10:327–336.
(Figure 5.13). One month later, this patient under- Ballarini et al. (2007). The physics of the FLUKA
went selective radioembolization of the remaining code: Recent developments. Adv Space Res
untreated lesions in the anterior right lobe. 40:1–11.
Bondiau, P.Y., Malandain, G., Chanalet, S. (2005).
Atlas-based automatic segmentation of MR
5.11 CONCLUSIONS images: Validation study on the brainstem in
radiotherapy context. Int J Radiat Oncol Biol
Treatment planning for radioembolization is not Phys 61:289–298.
only a multidisciplinary process requiring input Buie, H.R., Campbell, G.M., Klinck, R.J., MacNeil,
from different medical specialists but also a process J.A. (2007). Automatic segmentation of cortical
that requires significant experience. Differences and trabecular compartments based on a dual
between patients, even among those with the same threshold technique for in vivo micro-CT bone
disease type, can vastly effect the treatment decision analysis. Bone 41:505–515. Epub 2007 Jul 18.
and planning process. These differences necessitate Campbell, A.M., Bailey, I.H., Burton, M.A. (2000).
a patient-specific approach to treatment planning. Analysis of the distribution of intra-arterial
This chapter is but a brief overview of the fac- microspheres in human liver following hepatic
tors that must be considered by the interventional yttrium-90 microsphere therapy. Phys Med
radiologists, medical physicists, nuclear medicine Biol 45:1023–1033.
physicians, and/or radiation oncologists involved in Campbell, A.M., Bailey, I.H., Burton, M.A. (2001).
the therapy. In addition to the instruction provided Tumour dosimetry in human liver following
in this chapter, the most important suggestion that hepatic yttrium-90 microsphere therapy.
the authors wish to convey is the importance of Phys Med Biol 46:487–498.
110 Treatment planning part III

Caon, M. (2004). Voxel-based computational Garin, E., Boucher, E., Rolland, Y. (2014). 99mTc-
models of real human anatomy: A review. MAA-Based dosimetry for liver cancer
Radiat Environ Biophys 42:229–235. Epub treated using 90Y-loaded microspheres:
2004 Jan 17. Known proof of effectiveness. J Nucl Med
Carretero, C., Munoz-Navas, M., Betes, M. et al. 55:1391–1392.
(2007). Gastroduodenal injury after radioembo- Ghanei, A., Soltanian-Zadeh, H., Ratkewicz,
lization of hepatic tumors. Am J Gastroenterol A. (2001). A three-dimensional deformable
102:1216–1220. model for segmentation of human pros-
Cremonesi, M., Ferrari, M., Bartolomei, tate from ultrasound images. Med Phys
M. et al. (2008). Radioembolisation with 28:2147–2153.
90Y-microspheres: Dosimetric and radiobio- Giammarile, F., Bodei, L., Chiesa, C. et al. (2011).
logical investigation for multi-cycle treatment. EANM procedure guideline for the treatment
Eur J Nucl Med Mol Imaging 35:2088–2096. of liver cancer and liver metastases with intra-
Dawson, L.A., Guha, C. (2008). arterial radioactive compounds. Eur J Nucl
Hepatocellular carcinoma: Radiation Med Mol Imaging 38:1393–1406.
therapy. Cancer J 14:111–116. doi: 10.1097/ Goin, J.E., Salem, R., Carr, B.I. et al. (2005).
PPO.0b013e31816a0e80. Treatment of unresectable hepatocellular
Eckerman, K.F., Westfall, R.J., Ryman, J.C., carcinoma with intrahepatic yttrium 90 micro-
Cristy, M. (1994). Availability of nuclear spheres: Factors associated with liver toxici-
decay data in electronic form, including beta ties. J Vasc Intervent Radiol 16:205–213.
spectra not previously published. Health Phys Guha, C., Kavanagh, B.D. (2011). Hepatic radia-
67:338–345. tion toxicity: Avoidance and amelioration.
El-Baz, A.S., U, R.A., Mirmehdi, M., Suri, Semin Radiat Oncol 21:256–263.
J.S. (2011). Multi-Modality State-of-the-Art Gulec, S.A., Mesoloras, G., Dezarn, W.A. et al.
Medical Image Segmentation and Registration (2007). Safety and efficacy of Y-90 micro-
Methodologies. New York, NY: Springer sphere treatment in patients with primary and
Science and Business Media. metastatic liver cancer: The tumor selectivity
Emami, B., Lyman, J., Brown, A. et al. (1991). of the treatment as a function of tumor to
Tolerance of normal tissue to therapeu- liver flow ratio. J Transl Med 5:15.
tic irradiation. Int J Radiat Oncol Biol Phys Gulec, S.A.S., Mesoloras, G.G., Stabin,
21:109–122. M.M. (2006). Dosimetric techniques in
Foruzan, A.H., Zoroofi, R.A., Hori, M., Sato, 90Y-microsphere therapy of liver cancer: The
Y. (2009). Liver segmentation by intensity MIRD equations for dose calculations. J Nucl
analysis and anatomical information in multi- Med 47:1209–1211.
slice CT images. Int J Comput Assist Radiol Ho, S., Lau, W.Y., Leung, T.W. et al. (1996).
Surg 4:287–297. Partition model for estimating radiation
Fox, R.A., Klemp, P.F., Egan, G. et al. (1991). doses from yttrium-90 microspheres in
Dose distribution following selective internal treating hepatic tumours. Eur J Nucl Med
radiation therapy. Int J Radiat Oncol Biol Phys 23:947–952.
21:463–467. Ho, S.S., Lau, W.Y.W., Leung, T.W.T. et al. (1997).
François, C.J., Fieno, D.S., Shors, S.M., Finn, Clinical evaluation of the partition model for
J.P. (2004). Left ventricular mass: Manual estimating radiation doses from yttrium-90
and automatic segmentation of true FISP microspheres in the treatment of hepatic
and FLASH cine MR images in dogs and cancer. Eur J Nucl Med 24:293–298.
pigs. Radiology 230:389–395. Epub 2003 ICRU. (1992). Photon, Electron, Proton and
Dec 29. Neutron Interaction Data for Body Tissues.
Fripp, J., Crozier, S., Warfield, S.K., Ourselin, Bethesda, MD: International Commission on
S. (2007). Automatic segmentation of articular Radiological Units and Measurements.
cartilage in magnetic resonance images of Jiang, M., Fischman, A., Nowakowski, F.S.
the knee. Med Image Comput Comput Assist et al. (2012). Segmental perfusion differ-
Interv 10:186–194. ences on paired Tc-99m macroaggregated
References 111

albumin (MAA) hepatic perfusion imag- Lam, M.G.E.H., Goris, M.L., Iagaru, A.H. et al.
ing and yttrium-90 (Y-90) bremsstrahlung (2013). Prognostic utility of 90Y radioemboli-
imaging studies in SIR-sphere radioem- zation dosimetry based on fusion 99mTc-mac-
bolization: Associations with angiog- roaggregated albumin-99mTc-sulfur colloid
raphy. J Nucl Med Radiat Ther 3 :122. SPECT. J Nucl Med 54:2055–2061.
doi:10.4172/2155–9619.1000122. Lam, M.G.E.H., Smits, M.L.J. (2013). Value of
Jones, A.K., Mahvash, A. (2012). Evaluation of the 99mTc-macroaggregated albumin SPECT for
potential utility of flat panel CT for quantify- radioembolization treatment planning. J Nucl
ing relative contrast enhancement. Med Phys Med 54:1681–1682.
39:4149–4154. Lam, M.G.E.H., Sze, D.Y. (2014). Reply: 99mTc-
Kao, Y.H., Hock Tan, A.E., Burgmans, M.C. et al. MAA-based dosimetry for liver cancer
(2012). Image-guided personalized predic- treated using 90y-loaded microspheres:
tive dosimetry by artery-specific SPECT/ Known proof of effectiveness. J Nucl Med
CT partition modeling for safe and effec- 55:1392–1393.
tive 90Y radioembolization. J Nucl Med Lanconelli, N., Pacilio, M., Meo Lo, S. et al. (2012).
53:559–566. A free database of radionuclide voxel S values
Kennedy, A., Dezarn, W., Weiss, A. (2011). Patient for the dosimetry of nonuniform activity dis-
specific 3D image-based radiation dose esti- tributions. Phys Med Biol 57:517–533.
mates for 90Y microsphere hepatic radioem- Lau, W.Y., Leung, W.T., Ho, S. et al. (1994).
bolization in metastatic tumors. J Nucl Med Treatment of inoperable hepatocellular car-
Radiat Ther 2:1–8. cinoma with intrahepatic arterial yttrium-90
Kennedy, A.S. et al. (2009). Treatment param- microspheres: A phase I and II study. Br J
eters and outcome in 680 treatments of Cancer 70:994–999.
internal radiation with resin 90Y-microspheres Lewandowski, R.J., Thurston, K.G., Goin,
for unresectable hepatic tumors. Int J Radiat J.E. et al. (2005). 90Y microsphere
Oncol Biol Phys 74:1494–1500. (TheraSphere) treatment for unresect-
Kennedy, A.S.A., Nutting, C.C., Coldwell, able colorectal cancer metastases of the
D.D. et al. (2004). Pathologic response and liver: Response to treatment at targeted
microdosimetry of 90Y microspheres in man: doses of 135–150 Gy as measured by
Review of four explanted whole livers. Int J [18F]fluorodeoxyglucose positron emis-
Radiat Oncol Biol Phys 60:12. sion tomography and computed tomo-
Klein, S., van der Heide, U.A., Lips, I.M., graphic imaging. J Vasc Interv Radiol
van Vulpen, M. (2008). Automatic segmenta- 16:1641–1651.
tion of the prostate in 3D MR images by atlas Linguraru, M.G., Sandberg, J.K., Li, Z. et al.
matching using localized mutual information. (2010). Automated segmentation and quan-
Med Phys 35:1407–1417. tification of liver and spleen from CT images
Knesaurek, K., Machac, J., Muzinic, M. et al. using normalized probabilistic atlases
(2010). Quantitative comparison of yttrium-90 and enhancement estimation. Med Phys
(90Y)-microspheres and technetium- 37:771–783.
99m (99mTc)-macroaggregated albumin Louie, J.D., Kothary, N., Kuo, W.T. et al. (2009).
SPECT images for planning 90Y therapy of liver Incorporating cone-beam CT into the treat-
cancer. Technol Cancer Res Treatm 9:253–261. ment planning for yttrium-90 radioemboliza-
Kockelkorn, T.T.J.P., Schaefer-Prokop, C.M., tion. J Vasc Interv Radiol 20:606–613.
Bozovic, G. et al. (2014). Interactive Marroquín, J.L., Vemuri, B.C., Botello, S.
lung segmentation in abnormal human et al. (2002). An accurate and efficient
and animal chest CT scans. Med Phys Bayesian method for automatic segmenta-
41:081915–081915. tion of brain MRI. IEEE Trans Med Imaging
Kritzinger, J., Klass, D., Ho, S. et al. (2013). 21:934–945.
Hepatic embolotherapy in interventional McKinney, G.W., Durkee, J.W., Hendricks, J.S.
oncology: Technology, techniques, and appli- (2006). MCNPX overview. Los Alamos, NM:
cations. Clin Radiol 68:1–15. Los Alamos National Laboratory.
112 Treatment planning part III

Metzger, M.C., Bittermann, G., Dannenberg, South, C.D., Meyer, M.M., Meis, G. et al.
L. et al. (2013). Design and development of (2008a). W1672 yttrium-90 microsphere
a virtual anatomic atlas of the human skull Induced gastrointestinal tract ulceration.
for automatic segmentation in computer- Gastroenterology 134:A-904.
assisted surgery, preoperative planning, and South, C.D., Meyer, M.M., Meis, G. et al.
navigation. Int J Comput Assist Radiol Surg (2008b). Yttrium-90 microsphere induced
8:691–702. gastrointestinal tract ulceration. World J Surg
Naymagon, S., Warner, R.R.P., Patel, K. et al. Oncol 6:93.
(2010). Gastroduodenal ulceration associated Stabin, M.G., Eckerman, K.F., Ryman, J.C. et al.
with radioembolization for the treatment (1994). Bremsstrahlung radiation dose in
of hepatic tumors: An institutional experi- yttrium-90 therapy applications. J Nucl Med
ence and review of the literature. Dig Dis Sci 35:1377–1380.
55:2450–2458. Strigari, L., Menghi, E., D’Andrea, M., Benassi,
Park, S.H., Gao, Y., Shi, Y., Shen, D. (2014). M. (2006). Monte Carlo dose voxel kernel
Interactive prostate segmentation using atlas- calculations of beta-emitting and Auger-
guided semi-supervised learning and adap- emitting radionuclides for internal dosimetry:
tive feature selection. Med Phys 41:111715. A comparison between EGSnrcMP and EGS4.
Pasciak, A.S., Bourgeois, A.C., McKinney, Med Phys 33:3383.
J.M. et al. (2014). Radioembolization and Strigari, L., Sciuto, R., Rea, S. et al. (2010). Efficacy
the dynamic role of (90)Y PET/CT. Front and toxicity related to treatment of hepa-
Oncol 4:38. tocellular carcinoma with 90Y-SIR spheres:
Reed, V.K., Woodward, W.A., Zhang, L. et al. Radiobiologic considerations. Journal of
(2009). Automatic segmentation of whole nuclear medicine: Official publication. Soc
breast using atlas approach and deformable Nucl Med 51:1377–1385.
image registration. Int J Radiat Oncol Biol van Ginneken, B., Haar Romeny ter, B.M.
Phys 73:1493–1500. (2000). Automatic segmentation of
Riaz, A., Gates, V.L., Atassi, B. et al. (2011). lung fields in chest radiographs. Med Phys
Radiation segmentectomy: A novel approach 27:2445.
to increase safety and efficacy of radio- Warfield, S.K., Zou, K.H., Wells, W.M. (2004).
embolization. Int J Radiat Oncol Biol Phys Simultaneous truth and performance level
79:163–171. estimation (STAPLE): An algorithm for the
Salem, R., Thurston, K.G. (2006). validation of image segmentation. IEEE Trans
Radioembolization with 90Yttrium micro- Med Imaging 23:903–921.
spheres: A state-of-the-art brachytherapy Wondergem, M., Smits, M.L.J., Elschot, M.
treatment for primary and secondary liver et al. (2013). 99mTc-macroaggregated
malignancies. Part 1: Technical and meth- albumin poorly predicts the intrahepatic
odologic considerations. J Vasc Interv Radiol distribution of 90Y resin microspheres in
17:1251–1278. hepatic radioembolization. J Nucl Med
Sangro, B., Gil-Alzugaray, B., Rodriguez, J. et al. 54:1294–1301.
(2008). Liver disease induced by radioembo- Yoo, H.S., Park, C.H., Suh, J.H. et al. (1989).
lization of liver tumors: Description and pos- Radioiodinated fatty acid esters in the
sible risk factors. Cancer 112:1538–1546. management of hepatocellular carcinoma:
Sempoux, C., Horsmans, Y., Geubel, A. et al. Preliminary findings. Cancer Chemother
(1997). Severe radiation-induced liver disease Pharmacol 23(Suppl):S54–S58.
following localized radiation therapy for bil- Zhang, L.Z.L., Hoffman, E.A., Reinhardt,
iopancreatic carcinoma: Activation of hepatic J.M. (2006). Atlas-driven lung lobe segmenta-
stellate cells as an early event. Hepatology tion in volumetric X-ray CT images. IEEE Trans
26:128–134. Med Imaging 25:1–16.
6
Radioembolization in segmentectomy,
lobectomy, and future liver remnant
hypertrophy

EDWARD KIM, JOSEPH TITANO, AND SAFET LEKPERIC

6.1 Introduction 113 6.5 Radioembolization toxicities and


6.2 Treatment planning and delivery 114 complications 122
6.3 Radiation lobectomy and future liver 6.5.1 Radiation segmentectomy
remnant hypertrophy 114 toxicities 123
6.3.1 Definition and treatment 6.6 Posttreatment patient management 123
rationale 114 6.6.1 Patient care in the immediate
6.3.2 Radiation biology and postprocedure setting 123
radiation lobectomy 115 6.6.2 Follow-up evaluation 124
6.3.3 Patient selection 116 6.6.3 Imaging response 124
6.3.4 Lobectomy dosimetry 116 6.7 Clinical case examples 124
6.3.5 Radiation lobectomy and 6.7.1 Sample radiation lobectomy
the future liver remnant and the future liver remnant
hypertrophy outcome data 117 hypertrophy case 124
6.4 Radiation segmentectomy 119 6.7.2 Sample radiation
6.4.1 Definition and treatment rationale 119 segmentectomy case 126
6.4.2 Patient selection 120 6.8 Conclusions 128
6.4.3 Segmentectomy dosimetry 120 References 128
6.4.4 Radiation segmentectomy
outcome data 120

threefold intention of treating the tumor, allowing


6.1 INtrODUCtION a biological test of time to select for less aggressive
lesions and causing left lobe hypertrophy as a means
As the role of 90Y transarterial radioembolization of enabling right lobe surgical resection. The second
has evolved, specific treatment paradigms have led analogous concept is “radiation segmentectomy,”
to the development of radioembolization appli- in which a large dose of radiation is delivered to a
cations analogous to surgical liver interventions. small volume of liver thereby imparting a highly
The first such application is “radiation lobectomy,” tumoricidal dose to the perfused target, while spar-
in which radioembolization is performed to treat ing adjacent and nontarget liver parenchyma. These
an unresectable right lobe liver lesion with the concepts have broadened the armament physicians

113
114 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

have available to treat liver tumors by allowing for artery. Radioembolization is then generally per-
treatment of lesions with complex anatomic loca- formed through a coaxial 0.0325-inch system
tions and by expanding the patient population eligi- within the target vessel. During infusion, care must
ble for interventions. The radiation biology, physics, be taken to avoid stasis and reflux of the 90Y micro-
nuclear medicine, and interventional radiology spheres in order to avoid nontarget embolization,
concepts related to these treatment entities are dis- which could result in gastrointestinal ulceration,
cussed in this chapter. lung parenchymal injury, or pancreatitis among
other complications (Salem and Thurston, 2006c).
Following radioembolization completion and
catheter removal, postprocedure imaging with
6.2 trEatMENt PLaNNING single-photon emission computed tomography
aND DELIVErY (SPECT)/computed tomography (CT) or posi-
tron emission tomography (PET)/CT/magnetic
As described in Chapter 4, once a patient is selected resonance imaging (MRI) may be obtained to
for radioembolization therapy, treatment planning evaluate for nontarget deposition of 90Y micro-
begins with the completion of a technetium-99m spheres or to assess the distribution of micro-
macroaggregated albumin (99mTc-MAA) study. The spheres within the liver and tumor, respectively.
purpose of this study, again, is multifaceted. First, it Recently, 90Y PET/CT has gained increasing
is undertaken to visualize the vascular supply to the popularity compared with 90Y-bremsstrahlung
liver and to the lesion of interest. This allows for the SPECT/CT on account of its greater dosim-
potential embolization of vessels that have the poten- etry accuracy (Braat et al., 2015). While further
tial to divert the glass or resin 90Y microspheres away study is still required, delivered-dose calculation
from the intended target thereby causing adverse on posttreatment imaging may predict tumor
events. In addition, it allows for calculation of the response and thereby allow for early planning
lung shunt fraction (Uliel et al., 2012). of repeat interventions (Braat et al., 2015). More
Safe and effective delivery of radioemboliza- detailed discussion of these modalities will be
tion therapy—whether for lobar or segmental provided in Chapters 10 and 11.
infusion—requires careful evaluation in the inter-
ventional suite as well as the patient. Detailed
discussion of radiation safety will be provided in
6.3 raDIatION LOBECtOMY
Chapter 7. Briefly, the interventional suite should be
outfitted with radiation detection equipment such
aND FUtUrE LIVEr
that a thin-window Geiger–Müller counter that is rEMNaNt HYPErtrOPHY
able to detect radiation levels under 0.1 mR/hour is
available to detect the contamination of personnel, 6.3.1 DEFINITION AND TREATMENT
garbage, and the interventional suit equipment. RATIONALE
In addition, an ionization chamber able to detect
radiation doses of 1 mrem/hour should be avail- Radiation lobectomy, as its name implies, entails
able to localize the dose delivery site and measure lobar infusion of 90Y microspheres. Future liver
activity remaining in the dose vial. A large drape remnant (FLR) hypertrophy specifically refers to
should be prepared in close proximity to the fluo- right lobar delivery of 90Y microspheres in patients
roscopy table so that potential leaks are contained with right-sided tumors who would be candidates
immediately. An acrylic desiccator is also required for resection if the FLR were adequate. Adequate
in order to house the dose vile, tubing, and catheter volumes for the FLR have been cited between 20%
following radioembolization (Salem and Thurston, and 40% of total liver volume, with a larger rem-
2006c). nant recommended for cirrhotic patients (Kubota
Following standard prepping and draping, arte- et al., 1997; Zorzi et al., 2007; Shindoh et al., 2013;
rial access is gained—typically via the common Vouche et al., 2013). The intention behind radia-
femoral artery. A 4-French or 5-French catheter sys- tion lobectomy is threefold: (1) treat the right-sided
tem is generally utilized to navigate the aorta and to tumor, (2) simultaneously induce left liver lobe
cannulate the celiac axis or the superior mesenteric hypertrophy such that an adequate FLR is achieved
6.3 Radiation lobectomy / 6.3.2 Radiation biology and radiation lobectomy 115

allowing the patient to proceed with potentially FLR hypertrophy achieved by the combination of
curative procedures such as surgical resection, and TACE and PVE is equivalent to or arguably infe-
(3) allow for a test of time to identify less aggressive rior to that of PVE alone (Teo et al., 2015).
tumors in the hopes of limiting recurrence rates
postresection (Gaba et al., 2009; Inarrairaegui 6.3.2 RADIATION BIOLOGY AND
et al., 2012; Salem et al., 2013). RADIATION LOBECTOMY
Portal vein embolization (PVE) is an alterna-
tive to radiation lobectomy that is also employed Before examining the details of liver parenchymal
with the intention of inducing lobar hypertrophy change following radiation lobectomy, a general dis-
in order to produce an adequate FLR. While PVE cussion of liver tissue response to insult—and spe-
is perhaps even more effective than radioemboli- cifically radiation-induced insult—is warranted. A
zation (Azoulay et al., 2000; Pamecha et al., 2009; more robust discussion of radiation biology can be
Garlipp et al., 2014) at increasing the FLR volume, found in Chapters 8 and 9. Following parenchymal
there are several advantages offered by radiation injury, hepatic stellate cells migrate to the affected
lobectomy. First, PVE does not directly treat the region and begin to produce extracellular matrix
liver tumor; while it interrupts portal flow to the leading to fibrosis (Clement et al., 1986; Jakobs
lesion, the arterial supply from which tumors draw et al., 2008). As shown in studies primarily focused
the majority of their nutrition remains intact. This on external beam radiation, once the radiation
means that lesions remain unchecked while await- dose applied to liver tissue exceeds 30–40 Gy addi-
ing the FLR hypertrophy following PVE. Radiation tional pathological and morphological changes
lobectomy is also a microembolic therapy causing consistent with veno-occlusive disease (VOD)
radiation-induced atrophy of the target lobe allow- are seen including sinusoidal congestion, hemor-
ing for a delayed diversion of portal blood flow rhage, atrophy, and necrosis (Fajardo and Colby,
from the right lobe to the left lobe, which may allow 1980; Jakobs et al., 2008). The implication, then,
for a greater accommodation of increased blood is that radioembolization may induce a degree of
flow by the FLR (Jakobs et al., 2008; Gaba et al., portal hypertension (Jakobs et al., 2008; Gaba et
2009; Vouche et al., 2013). This pattern of hyper- al., 2009). The restorative mechanisms associated
trophy allows for a test-of-time through which with hepatic parenchymal insult are also initiated
tumor response to therapy may be assessed and following radioembolization. Hepatocyte prolif-
those patients with positive tumor biology may eration is the end result of multiple inputs directed
then be selected for further curative interventions by cytokines, growth factors, signaling pathway
(Vouche et al., 2013). Finally, the microembolic cascades, and transcription factor activation (Gaba
nature of radiation lobectomy allows for expansion et al., 2009).
of the treatment population to patients with portal While local veno-occlusive changes follow
vein thrombus (PVT). While resection of tumors radioembolization, radiation lobectomy does not
associated with PVT is rare in the United States, necessarily induce portal hypertension globally.
these resections are performed frequently in Asian In a study by Jakobs et al. (2008), in which volu-
hospital centers (Vouche et al., 2013). metric changes following radioembolization were
The combination of transarterial chemoemboli- explored, a subgroup analysis of those patients
zation (TACE) and PVE has been offered to account who underwent unilateral treatment of the right
for the lack of tumor control offered by PVE alone. liver lobe showed a significant increase in left liver
While one might expect that embolization of the lobe volume, a significant decrease in right liver
portal and arterial vessels supplying the same lobe volume, increased left portal vein diameter,
region of liver parenchyma may be associated with and no change in splenic volume. Splenic volumes
hepatic injury, increased toxicity is only transient were significantly increased in patients who under-
(Aoki et al., 2004). In addition to safety, sequen- went bilobar radioembolization (Jakobs et al.,
tial TACE and PVE have been shown to increase 2008). Together, these findings suggest that portal
the rate of the FLR hypertrophy, improve recur- venous flow is diverted into the left-sided portal
rence-free survival, and increase overall survival system—as evidenced by the increased diameter
in patients with hepatocellular carcinoma (HCC) of the left portal vein—without the development
(Ogata et al., 2006; Yoo et al., 2011). However, the of secondary signs of portal hypertension such as
116 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

an increase in splenic volume following right lobar and necrosis while the tumor itself is also treated.
radioembolization. The veno-occlusive changes that follow radioem-
Recently, Fernandez-Ros et al. (2015) described bolization then lead to a slow redirection of por-
in detail the mechanisms of the biological response tal vein blood flow to the untreated left liver lobe.
to radioembolization specifically. They describe Simultaneously then, signaling pathways are initi-
oxidative stress as the driver behind endothelial cell ated and proliferative mediators are recruited while
injury as well as activation of coagulation and pro- portal venous flow is directed toward the FLR (Gaba
inflammatory pathways. While it remains unclear et al., 2009; Vouche et al., 2013).
if the elevation of proinflammatory markers is a
primary effect of radioembolization or follows 6.3.3 PATIENT SELECTION
secondarily from VOD, the role of coagulation in
VOD (as shown in the pathologically similar VOD Generally, radioembolization is indicated for
following bone marrow transplantation) has been patients with unresectable HCC, cholangiocar-
better established (Fernandez-Ros et al., 2015). cinoma, or with metastatic liver lesions. In 1999,
At a cellular level, the changes that have been the U.S. Food and Drug Administration (USFDA)
described following portal vein ligation and partial issued a humanitarian device exemption for glass
hepatic resection have shown that hepatocytes pro- microspheres as neoadjuvant therapy prior to sur-
liferate initially followed by nonparenchymal cells gery or transplantation in patients with unresectable
(Michalopoulos and DeFrances, 1997; Fernandez- HCC. Similarly, in 2002, the USFDA approved the
Ros et al., 2015). In addition to the redistribution use of 90Y resin microspheres for the treatment of
of portal blood flow, mitogens including hepato- unresectable metastatic liver tumors from primary
cyte growth factor (HGF), fibroblast growth fac- colorectal cancer with adjuvant intrahepatic artery
tor type 19 (FGF-19), interleukin 6 (IL-6), and chemotherapy.
insulin are also increased following these proce- Against this background, radiation lobectomy
dures. Specifically following radioembolization, is a specific application of radioembolization in
significant increases in HGF and FGF-19 are noted patients with hepatic tumor lesions that would be
(Fernandez-Ros et al., 2015). Both of these factors eligible for definitive therapy with hepatic lobar
drive transcription factor activation and initiate resection if the remaining FLR were adequate (Gaba
hepatocyte regeneration. In addition, IL-6 and et al., 2009; Siddiqi and Devlin, 2009; Vouche et al.,
tumor necrosis factor-alpha (TNF-α) also drive 2013). Radiation lobectomy allows for the treatment
hepatocyte replication by initiating the transition of right hepatic lobe tumor burden while inducing
of these cells from G0 to G1 (Fernandez-Ros et the FLR hypertrophy through the redirection of
al., 2015). Sustained increases in TNF-α and IL-6 portal blood flow and by the production of growth
are observed following treatment and likely con- factors and cytokines. In addition, the time interval
tribute to the FLR hypertrophy (Fernandez-Ros required to allow for future remnant hypertrophy
et al., 2015). In contrast to TACE following which mandates a period prior to surgical intervention
transient increases in IL-6 and HGH have been that allows for aggressive tumors to declare them-
documented, these factors were noted to be ele- selves on follow-up imaging (Vouche et al., 2013).
vated months after radioembolization—a timeline
commensurate with that of the FLR hypertrophy 6.3.4 LOBECTOMY DOSIMETRY
(Yamazaki et al., 1996; Kim et al., 2013; Fernandez-
Ros et al., 2015). This contrast with TACE also pro- Generally, dose calculation for radiation lobec-
vides a basis for the suggestion that IL-6 and HGH tomy and the FLR hypertrophy is performed by
sustained elevations are more likely the result of completing calculations for lobar therapy as dis-
radiation effects than embolic effects of therapy cussed in Chapter 5. Briefly, under assumptions of
(Fernandez-Ros et al., 2015). uniform dose distribution and complete 90Y decay
Radiation lobectomy, then, harnesses multiple as elaborated upon in the previous chapter, the
processes leading to both atrophy and hypertrophy administered activity is calculated using Equation
contributing to the therapeutic aim of increasing the 5.9 where Ao is the treatment activity, Davg is the
volume of the FLR. The treated right lobe undergoes desired average absorbed dose, and Mliver is the
the development of sinusoidal congestion, atrophy, mass of the liver to be treated:
6.3 Radiation lobectomy / 6.3.5 Radiation lobectomy and FLR hypertrophy outcome data 117

Davg (Gy) ⋅ M liver (kg) resonance imaging (SHARP or VIBE sequences) or


Ao (GBq) = computed tomography. Boundaries for the right lobe
49.98 (J ⋅ s)
and the left lobe were delineated by the left hepatic
Again, the lobar mass is utilized as the mass of vein in the upper lobe and a line drawn from the
liver intended to undergo treatment in this dose inferior vena cava to the insertion of the falciform
calculation paradigm. The mass of the liver to be ligament in the lower lobes. The portal triad, gall-
treated, Mliver, is obtained by measuring the target bladder, and inferior vena cava were excluded from
liver volume and converting the volume measure volumetric analysis. According to this system, the
to a calculated mass value 1.05 kg/L. right liver lobe volume consists of the combined
Following treatment, the actual dose delivered measures of segments 1, 4, 5, 6, 7, and 8 while the left
may be determined utilizing Equation 6.1: liver lobe volume consists of the combined measures
of segments 2 and 3.
Ao (GBq ) (1 − SF )(1 − R ) ⋅ 49.98 (J ⋅ s)
Ddelivered (Gy ) =  While usage of the term FLR has been defined
M liver (kg) in several ways, we have selected to define it as the
(6.1) percentage of the FLR volume as a ratio of total
liver volume (Vouche et al., 2013). Integral to the
where SF is the lung shunt fraction and R is the
discussion of liver volume changes following radi-
percentage of dose remaining within the vial at the
ation lobectomy are the following equations:
completion of treatment. Currently, there is no uni-
versal dosing pattern for radiation lobectomy and LLPV
the FLR hypertrophy. The dosing information in the FLR% =   ⋅ 100% (6.2)
TLPV
major studies of radiation lobectomy is provided in
Table 6.1. In the largest study of resin microspheres
%FLR Hypertrophy
utilized for radiation lobectomy and the FLR hyper-
trophy, Fernandez-Ros et al. (2014) found no cor- FLR post-Y90 – FLR pre-Y90
= ⋅ 100%, (6.3)
relation between dose and volume changes and FLR pre-Y90
concluded that—while it is currently unknown
increased dosing could enhance the FLR hypertro- where LLPV is defined as the volume of segments 2
phy—hypertrophy does indeed occur at therapeutic and 3 less the total volume of tumor within the left
doses. Similarly, in the largest study of glass micro- lobe, and TLPV is defined as the total liver volume
spheres used for radiation lobectomy and the FLR less the volume of total tumor burden within the
hypertrophy, Vouche et al. (2013) reported a median liver (Vouche et al., 2013).
dose of 112 Gy (range: 74–215 Gy) delivered to the
treatment site and included dose ≤100 Gy and dose
6.3.5.2 Liver volume changes,
>120 Gy as variables in their multivariate analysis of
%FLR hypertrophy (defined below), with neither of subsequent therapies, and
the conditions meeting statistical significance. survival outcomes
Several studies have evaluated the efficacy of radia-
6.3.5 RADIATION LOBECTOMY
tion lobectomy in producing the FLR hypertro-
AND THE FUTURE LIVER phy. The findings of these studies are provided in
REMNANT HYPERTROPHY Table 6.1. Jakobs et al. (2008) evaluated volumetric
OUTCOME DATA changes in the liver following lobar treatment with
90Y in a cohort predominantly of colorectal metas-

6.3.5.1 Imaging response tasis cases; a subanalysis of the those patients who
received unilateral right lobar treatment revealed
In the assessment of the FLR hypertrophy, volu- significantly decreased right lobe volume with sub-
metric measures of liver parenchyma are obviously sequent left lobe hypertrophy and no significant
essential. In the largest cohort of glass microsphere increase in spleen volume (Jakobs et al., 2008).
radiation lobectomy cases, Vouche et al. (2013) com- These findings contributed to the notion that
pleted computer-assisted volumetric assessment of radiation lobectomy allowed for a gradual, well-
the liver on either gadolinium-enhanced magnetic compensated diversion of portal venous flow from
Table 6.1 Radiation lobectomy and the FLR hypertrophy selected studies
Number time to Future
of Patient age Micro- Single or multiple activity/dose volume Imaging response Overall resection or
Study patients (years) tumor path spheres treatments information measure FLr hypertrophy summary survival transplantation

Ahmadza- 24 Median 63 CRC 15, Resin Staged lobar Mean activity 1.67 Mean 44 days, Mean 47%, median RECIST, r disease group NR One patient with
dehfar et al. (range breast 5, treatments 4–6 GBq, median median 36 34% right lobe CR 2, PR 14, unilobar CRC
(2013) 44–78) pancreatic weeks apart for activity 1.75 days PD 1; bilobar disease metastases
2, gastric 17 patients with GBq (range group left lobe PR 1, went on to
1, unknown bilobar disease; 0.40–3.90 SD 4, PD 12; unilobar right
primary 1 single treatment Gbq), dose NR group right lobe PR 4, hepatectomy
for 7 patients SD 2, PD 1
with unilobar
disease

Edeline et al. 34 NR HCC Glass 30, Single Median dose to 3 months Mean 29% at 3 mRECIST Median 13.5 NR
(2013) Resin 4 treated months CR 29.6%, PR 33.3%, SD months
segment 122.1 Mean 42% at all 29.6%, PD 7.4%
Gy (90.4–210.5 available time
Gy) points

Fernandez-Ros 83 Median 66 HCC 52, CRC Resin Single NR 4–26+ weeks Mean NR NR NR
et al. (2014) (IQR 13, IHC 4, approximately
53–79) Other 14 45%

Garlipp et al. 26 Mean 59.2 CRC 18, Resin Single Median activity 1.2 Median 46 Mean 29% RECIST NR NR
(2014) (standard Breast 8, GBq (range days (27–79 (standard CR 1, PR 19, SD 5, PD 1
deviation Other 6 0.8–1.7 GBq) days) deviation
11.1) Dose NR 22.9%), median
25.3%

Teo et al. 17 Median 72 HCC Resin Single NR Median 5 Mean 34.2% RECIST, CR 2, PR 5, SD 6, NR One patient
(2014) (42–78) months (standard PD 4 underwent
(range 2–12 deviation 34.9%; surgical right
months), range lobectomy
mean 5.7 19.0–106.5%)
months

Theysohn et al. 45 Mean 71.9 HCC Glass Single Mean dose to 1–12 months Mean 50.46% at 6 NR NR NR
(2014) (range right lobe 112 months, mean
55–90) Gy (range 56.49% at 12
100–160 Gy) months

Vouche et al. 83 Median 68 HCC 67, IHC Glass Single Median dose to 1–9+ months Median maximal NR Median Five patients
(2013) (range 8, CRC 8 treatment site FLR 26%, survival underwent
36–89) 112 Gy (range median 45% at BCLC B surgical right
74–215 Gy) > 9 months and C lobectomy; 6
(5–186) patients patients
118 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

was 34.4 underwent


and 9.6 OLT
months,
respectively

Note: CR, complete response; CRC, colorectal cancer; IQR, interquartile range; NR, not reported; PD, progressive disease; PR, partial response; SD, stable disease.
6.4 Radiation segmentectomy / 6.4.1 Definition and treatment rationale 119

the right lobe to the FLR without inducing global single treatment session (Rhee et al., 2005; Riaz
portal hypertension. et al., 2011). The term segmentectomy was utilized
A study by Gaba et al. (2009) provided similar in reference to the resultant atrophy of the treated
results in a cohort of HCC and cholangiocarci- segments seen at follow-up imaging, which is anal-
noma cases with statistically significant increases ogous to segmental surgical hepatic resection.
and decreases in left lobe and right lobe volumes, Several factors created the clinical need for
respectively, measured at an average of 18 months radiation segmentectomy. First, small tumors
posttreatment (Gaba et al., 2009). Similarly, (those ≤3 cm) are generally considered for cura-
Vouche et al. (2013) demonstrated statistically sig- tive therapies including transplantation, surgical
nificant lobar volume changes and showed a linear, resection, and ablation (Llovet et al., 1999). If, how-
time-dependent hypertrophy of the FLR. In their ever, a lesion is not amenable to curative interven-
cohort of 83 patients, 5 went on to lobar resection tion on account of anatomic considerations (e.g.,
and 6 underwent liver transplantation. Vouche et adjacent to the dome of the liver and diaphragm
al. (2013) also showed that volumetric changes are or in close proximity to large vessels), comorbidi-
apparent as early as 1 month posttreatment with ties, or inadequate functional liver reserve, radia-
maximum FLR hypertrophy achieved at approxi- tion segmentectomy remains a viable treatment
mately 9 months posttreatment. Interestingly, the option for these patients. Second, several authors
presence of portal vein thrombosis was a signifi- have shown that increased radiation dose is asso-
cant predictor of the FLR hypertrophy >40% with ciated with improved tumor response (Ben-Josef
the implication that existing portal vein throm- et al., 2005; Riaz et al., 2011; Vouche et al., 2014),
bus might act as a naturally occurring portal vein and radiation segmentectomy allows for greater
embolization with even earlier diversion of portal activity delivery directly to a target lesion. Further,
flow to the FLR (Vouche et al., 2013). it has also been theorized that lower radiation dose
In addition to portal vein thrombus predict- applied to normal hepatic parenchyma minimizes
ing increased FLR hypertrophy, a recent study by injury to the normal tissue allowing for greater
Teo et al. (2014) showed that HCC patients with physiologic regeneration of normal parenchyma
underlying hepatitis B may achieve greater FLR (Riaz et al., 2011).
hypertrophy than their counterparts with hepati- In direct comparison with ablative procedures,
tis C or alcoholic liver disease. This finding, com- there are several advantages and disadvantages to
bined with a trend observed by Fernandez-Ros et radiation segmentectomy. Advantages of radia-
al. (2014) toward reduced the FLR hypertrophy, tion segmentectomy include the obviation of per-
implies that cirrhosis may somewhat limit the ben- cutaneous needle and probe placement with the
efits of lobectomy in generating FLR hypertrophy. associated theoretical risk of tract seeding and the
Survival data for studies focused on radiation ability to target high-risk ablation lesions (Riaz
lobectomy and FLR hypertrophy specifically are et al., 2011; Vouche et al., 2014). Disadvantages
reported at up to a median of 36.6 months and of segmentectomy relative to ablative procedures
are in line with concurrently published prospec- potentially include cost and radiation exposure,
tive and retrospective cohorts (Gaba et al., 2009; although ablation probes are often placed with
Vouche et al., 2013). CT guidance making this a relative disadvantage
(Vouche et al., 2014).
In addition to its complimentary role with other
ablative therapies, radiation segmentectomy also
6.4 raDIatION has several advantages compared with external-
SEGMENtECtOMY beam radiation therapy. These advantages are
mainly linked to anatomic and practical consider-
6.4.1 DEFINITION AND TREATMENT ations regarding treatment planning and delivery.
RATIONALE Lesions within the caudate lobe and the dome of
the liver put adjacent structures such as the lung
Radiation segmentectomy is defined as radioem- parenchyma and porta hepatis ducts and vessels
bolization of two or fewer hepatic segments—as at increased risk (Riaz et al., 2011). In addition,
delineated by the Couinaud system—during a while dose fractionation has shown benefits in
120 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

targeting radiosensitive as well as resistant malig- to a tumor is complicated by the physiology of


nant cells, this therapeutic approach requires mul- blood flow to liver tumors and to normal hepatic
tiple treatment sessions (Riaz et al., 2011). A final parenchyma. First, the formulae applied for dose
practical consideration is that respiratory motion calculations often assume uniform distribution
potentially puts lung parenchyma at risk during of microspheres within the treated volume of
external-beam radiation delivery in a manner that liver. However, it has been shown through a num-
is avoided with transarterial delivery of radiation ber of modalities that blood flow is preferentially
with the maximum tissue penetration of 11 mm diverted toward tumor compared with normal
associated with 90Y (Salem and Thurston, 2006a; liver parenchyma (Lau et al., 1994; Ho et al., 1996;
Riaz et al., 2011). Campbell et al., 2000; Sarfaraz et al., 2003; Riaz
et  al., 2011). Intuitively, this matches an essential
6.4.2 PATIENT SELECTION tenant of transarterial liver tumor therapy that
liver tumors draw a majority of their blood sup-
Based on the definition of radiation segmentec- ply from the hepatic arterial system while normal
tomy, a lesion must be isolatable within only two parenchyma receives a majority of its blood supply
segments of the liver supplied by the hepatic arte- from the portal venous system.
rial vessel selected for delivery of 90Y microspheres. Attempts to account for the nonuniform distri-
As discussed above, segmentectomy is complimen- bution of blood flow—and therefore of 90Y micro-
tary to ablation in that suboptimal lesions for abla- spheres—have been made previously. Riaz et al.
tion may be treated by radiation segmentectomy. (2011) identified the problems associated with the
The most often cited reason that a lesion is deferred assumption of uniformity in microsphere distri-
for ablation is that a lesion is located at the dome of bution and sought to account for these issues by
the liver in close proximity to the diaphragm and incorporating a subjectively determined ratio of
lung tissue (Riaz et al., 2011). Additional anatomic tumor hypervascularity relative to adjacent normal
considerations leading to the choice of radiation liver tissue following a review of angiography and
segmentectomy over ablation include proximity cross-sectional imaging studies. Although not an
to vessels and biliary structures, caudate lobe loca- ideal means of quantifying the asymmetric distri-
tion, and proximity to small bowel, large bowel, bution of blood flow to tumor relative to surround-
the gallbladder, or the heart (Vouche et al., 2014). ing normal tissue, this method demonstrated that
Recent publications have also demonstrated the such differences in calculation lead to more than
safety and efficacy of radiation segmentectomy in doubling of the median calculated dose delivered
patients with moderate hepatic dysfunction and to tumor—from 521 Gy (95% CI: 404–645 Gy) to
advanced disease including portal vein invasion 1214 Gy (95% CI: 961–1546 Gy) in their cohort of
(Padia et al., 2014). 84 patients (Riaz et al., 2011).

6.4.3 SEGMENTECTOMY 6.4.4 RADIATION


DOSIMETRY SEGMENTECTOMY
OUTCOME DATA
Generally, dose calculation for radiation segmen-
tectomy is performed by completing calculations
intended for treatment of the entire lobe in which 6.4.4.1 Radiation segmentectomy
the lesion is located; however, intra-arterial injec- imaging response
tion of the lobar dose is performed from a seg-
mental vessel supplying one or two segments as Imaging response in the studies focused on the
described previously (Rhee et al., 2005; Vouche et methodology of radiation segmentectomy is sum-
al., 2014). Equation 5.9 may be utilized in order to marized in Table 6.2. Riaz et al. (2011) presented
calculate the activity to be delivered. imaging response in accordance with World
Following treatment, the actual dose deliv- Health Organization (WHO) and European
ered may be determined utilizing Equation 6.1. Association for the Study of the Liver (EASL)
However, accurate calculation of activity delivered guidelines. EASL response was reported in 81%
Table 6.2 Radiation segmentectomy selected studies
Number
of Patient age tumor Micro- Imaging response
Study patients (years) path spheres Dose Follow-up time summary Overall survival

Padia et al. 20 Median 61 HCC Glass Median dose to Median 275 Time to EASL response: 33 90% at median
(2014) (range segment 254 Gy days (range days (range 5–133 days). follow-up of 275
54–76) (range 105–1055 Gy), 32 –677 days) EASL: CR 19, SD 1 days (range
median dose to 32–677 days)
tumor 536 Gy (range
203–1618 Gy)
Rhee et al. 14 Mean 62 HCC Glass Median dose to Median 185 NR NR
(2005) (range segment 348 Gy days (range
41–78) (range 105–857 Gy) 35–600 days)
Riaz et al. 84 Median 68 HCC Glass Median dose to NR TTP 13.6 months (95% CI, Median overall
(2011) (43–90) segment 521 Gy 9.3–18.7 months); EASL: survival 26.9
(range 404–645 Gy) response in 81% of months (95% CI,
patients; median time to 20.5–30.2
response 1.2 months months)
(95% CI, 1.1–1.4 months);
WHO: response in 59% of
patients; median time to
response 7.2 months
(95% CI, 4.2–8.5 months)
Vouche et al. 102 Median 64 HCC Glass Median dose to Median 27.1 mRECIST: CR 47%, PR 39%, Median overall
(2014) (IQR, segment 242 Gy months SD 12%, PD 1% survival
58–74) (IQR, 173–369 Gy) uncensored 53.4
months; median
overall survival
censored for
transplantation
34.5 months
Note: CR, complete response; IQR, interquartile range; NR, not reported; PD, progressive disease; PR, partial response; SD, stable disease; TTP, time to progression; WHO, World Health Organization.
6.4 Radiation segmentectomy / 6.4.4 Radiation segmentectomy outcome data 121
122 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

of patients with median time-to-disease response liver dysfunction (e.g., Childs–Pugh score)—given
1.2 months, and WHO response was reported the inherent limitations in comparing locoregional
in 59% of patients with median time-to-disease therapy survival with that of ablation (Lencioni et al.,
response of 7.2 months. The largest series pub- 2005; Chen et al., 2006; Livraghi et al., 2008; Pompili
lished to date by Vouche et al. (2014) demonstrated et al., 2013; Vouche et al., 2014). Further, Vouche et
complete response in 47% of patients, partial al. (2014) postulate that radiation segmentectomy
response in 39% of patients, and stable disease in might offer similar survival outcomes as those seen
12% of patients according to modified Response in Barcelona Clinic Liver Cancer (BCLC) A patients.
Evaluation Criteria for Solid Tumors (mRECIST). While controversies exist regarding the limitations
Median time-to-disease progression in this series of percutaneous ablation, radiation segmentectomy
was 33.1 months with new intrahepatic lesions remains a viable option for complex lesions with the
responsible for disease progression in a major- potential for complete pathological necrosis of the
ity of cases (Vouche et al., 2014). For comparison, target lesion (Salem et al., 2015; Seror et al., 2015).
median time-to-disease progression has been
reported between 7.9 and 33.3 months in prospec- 6.4.4.3 Radiation segmentectomy
tive studies and large cohort retrospective stud- dose and response
ies (Lewandowski et al., 2009; Salem et al., 2010;
relationship
Sangro et al., 2011).
As the goal of radiation segmentectomy is to
6.4.4.2 Radiation segmentectomy achieve complete pathological necrosis (CPN)
survival outcomes equivalent to that seen in analogous ablation pro-
cedures, several authors have focused on the radia-
Median overall survival for patients undergo- tion dose necessary to achieve such an outcome. By
ing radiation segmentectomy has been reported utilizing 99mTc-MAA SPECT/CT as a surrogate in
at 13.6–53.4 months (Riaz et al., 2011; Vouche dosimetry calculations, Garin et al. (2012) estab-
et  al., 2014). Censored for transplantation, the lished a threshold dose of 205 Gy as predictive of
overall survival in the 34.5 months in the Vouche EASL imaging response, progression-free survival,
et  al. (2014) study compared with the uncen- and overall survival. Specific to radiation segmen-
sored median overall survival of 53.4 months. tectomy, in a study by Vouche et al. (2014), 33 of
These overall survival rates are comparable with the 102 patients studied went on to receive liver
or exceed overall survival reported in other large transplantation; explant analysis revealed that a
cohorts of patients undergoing radioembolization dose >190 Gy was associated with CPN. Their find-
(Sangro et al., 2011). The variability in the reported ings suggest that a threshold radiation dose exists
segmentectomy median overall survival is likely to achieve CPN. In their study of highly selec-
secondary to the duration of the largest studies of tive radiation segmentectomy, Padia et al. (2014)
radiation segmentectomy—5 and 8 years, respec- reported a median dose of 255 Gy (range 105–1055
tively (Riaz et al., 2011; Vouche et al., 2014). In Gy) with a complete EASL response noted in 19 of
addition, sorafenib was approved late in 2007 and 20 patients in their cohort. A summary of dose,
was therefore unavailable for approximately half imaging response, and survival outcomes for sev-
of the Riaz et al. (2011) study period, while it was eral radiation segmentectomy studies is included
available for the majority of the Vouche et al. (2014) in Table 6.2.
study duration (Llovet et al., 2008).
Because radiation segmentectomy is ideally
suited as a complimentary therapy to ablation in 6.5 raDIOEMBOLIZatION
instances wherein ablation options are limited,
tOXICItIES aND
comparison to overall survival in ablation studies
is warranted. Vouche et al. (2014) argue that over-
COMPLICatIONS
all survival as well as local control of tumor lesions
in cases of radiation segmentectomy does not dif- The most commonly encountered clinical adverse
fer dramatically from overall survival achieved by effect following radioembolization is fatigue
ablation when stratified by measure of baseline (Salem and Thurston, 2006a). In addition, patients
6.6 Posttreatment patient management / 6.6.1 Patient care 123

may experience fever and chills up to several days 6.5.1 RADIATION


following treatment—likely secondary to the SEGMENTECTOMY
effects of radiation on normal hepatic parenchyma TOXICITIES
and subsequent release of endogenous pyrogens
(Murthy et al., 2005; Salem et al., 2005; Salem and Of particular interest to radiation segmentectomy
Thurston, 2006c). Additional clinical toxicities is bilirubin toxicity as the concentration of a lobar
described include abdominal pain, nausea, vom- dose within one or two segments imposes the theo-
iting, anorexia, diarrhea, and weight loss (Riaz et retical risk of increased biliary complications (Riaz
al., 2011; Vouche et al., 2014);. While these adverse et al., 2011). REILD is less of a concern in radia-
effects are often self-limited, endoscopic evaluation tion segmentectomy compared with treatments
should be considered if symptoms are persistent as involving greater volumes of liver as the target
they may reflect the development of gastrointesti- liver parenchyma volume is reduced without limit-
nal ulceration (Salem and Thurston, 2006c). ing tumor treatment efficacy (Sangro et al., 2008).
Laboratory toxicities following radioemboliza- However, the increased radiation dose delivered to
tion most commonly include abnormalities in bili- a smaller volume of liver parenchyma raises con-
rubin, albumin, AST, ALT, alkaline phosphatase, cern for injury to the biliary system. Rhee et al.
platelet levels, lymphocyte counts, and interna- (2015) reported a statistically significant increase
tional normalized ratio (INR). Care must be taken in bilirubin following segmental infusion of a lobar
in the interpretation of laboratory toxicities as it is radiation dose but deemed this change clinically
often difficult to separate progressive liver disease insignificant as only one of the 14 patients included
secondary to underlying disease versus treatment in their study had a change in Childs–Pugh class
toxicity. In a majority of cases, laboratory abnor- following radioembolization (Rhee et al., 2005).
malities are of limited clinical significance and Riaz et al. (2011) reported 5% of patients within
occasionally reflect preexisting poor liver func- their cohort of 84 patients undergoing radiation
tion and cirrhosis (Rhee et al., 2005; Vouche et al., segmentectomy developed small bilomas postpro-
2014). cedure as well as 1% of patients developing biliary
Radioembolization-induced liver disease (REILD) stricture within the treated segment (Riaz et al.,
is analogous to the radiation-induced liver disease 2011). Vouche et al. (2014) did not report biloma or
(RILD) described in the external beam radiation biliary stricture development in their multicenter
therapy literature and characterized by jaundice, cohort of 102 patients. Padia et al. (2014) also did
fatigue, and the development of ascites typically not report biloma or biliary stricture in their 20
1–2 months posttreatment in the absence of tumor patient cohort. For comparison, in a study of 327
progression or bile duct obstruction (Lawrence patients who underwent standard lobar treatment
et al., 1995; Sangro et al., 2008). REILD represents protocols, Atassi et al. (2008) found 17 cases of
a form of VOD as does RILD; the two entities differ biliary necrosis, 8 cases of biliary strictures, and 3
in that RILD presentation is described as an “anic- cases of biloma.
teric ascites” with a proportional elevation of liver
enzymes in contrast to REILD, which is character-
ized by markedly increased bilirubin (Sangro et al.,
6.6 POSttrEatMENt PatIENt
2008). Risk factors for the development of REILD
have been identified as prior treatment with che-
MaNaGEMENt
motherapy, relative young age, elevated baseline
bilirubin, cirrhosis at baseline, whole-liver treat- 6.6.1 PATIENT CARE IN THE
ment (versus lobar therapy), and the ratio of activity IMMEDIATE POSTPROCEDURE
administered relative to the volume of treated liver SETTING
(Sangro et al., 2008; Riaz et al., 2009a; Fernandez-
Ros et al., 2015). Additional discussion related to Radioembolization is generally performed on an
the side effects of radioembolization can be found outpatient basis. Following treatment, patients
in other chapters in this book. Chapter 14 focuses recover for 2–6 hours prior to discharge home.
on identifying and managing clinical sequelae using Practice patterns vary between institutions, but
posttreatment imaging. patients may be sent home with gastrointestinal
124 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

ulcer prophylaxis such as proton pump inhibitors 6.6.3 IMAGING RESPONSE


or with steroid tapers for treatment of fatigue. All
patients require instruction regarding radiation Generally, follow-up imaging with contrast-
safety precautions as their body surface readings enhanced CT or MRI is obtained at 1–3 months
may reach 1 mrem/hour following radioembo- posttreatment and then at 3- to 6-month intervals
lization (Salem and Thurston, 2006c). Further, for lesion surveillance and future treatment plan-
patients treated with resin should be advised that ning regardless of whether radioembolization is
urinary excretion of radioactivity is possible at performed in segmental or lobar treatment. Optimal
trace levels (25–50 kBq per liter of urine per GBq imaging response is expected to be seen approxi-
of dose), which may be safely disposed of with mately 3–6 months following therapy with median
standard precautionary methods. While the toxic- time to response 6.6 months for change in size and
ity profile of radioembolization is limited, patients 1.2 months for evidence of necrosis (Sangro et al.,
should again be reminded of potential treatment 2006; Riaz et al., 2009b; Salem et al., 2010, 2013).
adverse effects that they may experience following
discharge.
6.7 CLINICaL CaSE EXaMPLES
6.6.2 FOLLOW-UP EVALUATION
Patients are typically seen in an outpatient setting
6.7.1 SAMPLE RADIATION
2 weeks following treatment with radioemboli- LOBECTOMY AND THE
zation. At the time of this assessment, providers FUTURE LIVER REMNANT
should focus their history and examination on HYPERTROPHY CASE
the preservation of the patient’s performance sta-
A 68-year-old male with a past medical history sig-
tus in addition to monitoring for adverse effects,
nificant for hepatitis C cirrhosis was screened for
tumor lysis syndrome, or toxicities of nontarget
liver cancer using gadolinium contrast-enhanced
organs such as the lungs and the gastrointestinal
magnetic resonance imaging that revealed a right
tract. Laboratory testing may be obtained at this
hepatic lobe, segment 8 HCC (Figure 6.1). The
point and is likely to show lymphopenia, transient
patient was scheduled for right radioembolization
increases in aminotransferase levels, and transient
lobectomy following a tumor board conference
increases in tumor markers (Salem and Thurston,
attended by transplant surgeons, hepatologists,
2006c). Follow-up imaging is obtained as described
medical oncologists, and interventional radi-
below.
ologists. Resin microspheres were utilized in this

(a) (b)

Figure 6.1 Magnetic resonance T1-weighted gadolinium-enhanced arterial phase (a) and portal
venous phase (b) sequences demonstrating a right liver lobe mass measuring >3 cm with early arterial
enhancement and portal venous washout consistent with hepatocellular carcinoma.
6.7 Clinical case examples / 6.7.1 Sample radiation lobectomy and the future liver remnant hypertrophy case 125

patient’s radioembolization lobectomy procedure. Absa = 1.96 GBq


The activity calculations are provided following
the body surface area method presented in Chapter If glass microspheres had been utilized in this
5—specifically Equations 5.6 and 5.7: radiation lobectomy case, a sample of the hypo-
thetical activity calculation is provided utilizing
( )
BSA m 2 = 0.2025 ⋅ height 0.725 (m) ⋅ weight 0.425 (kg) Equation 5.9 following the conversion of the lobar
volume to mass (conversion factor of 1.05 kg/L):
( )
BSA m 2 = 0.2025 ⋅ 1.880.725 ⋅ (87.1)0.425
Davg  (Gy ) ⋅ M liver (kg)
Ao  (GBq ) = 
( ) 2
BSA m = 0.2025 ⋅ 1.58 ⋅ 6.68 49.98 (J ⋅ s)

BSA = 2.13 m 2  120 Gy ⋅ 1.42 kg


Ao  (GBq ) = 
Vtumor 49.98 (J ⋅ s)
Absa (GBq ) = BSA − 0.2 + 
Vtumor + Vnormal Ao = 3.42 GBq
38.8 mL
Absa (GBq ) = 2.13 − 0.2 + 
38.8 mL + 1381.2mL Following successful radioembolization of
the right lobe, subsequent right hepatic lobe
Absa (GBq ) = 2.13 − 0.2 + 0.03 atrophy and compensatory left hepatic lobe

(a) (b)

(c) (d)

Figure 6.2 Magnetic resonance T1-weighted precontrast (a), T1-weighted gadolinium-enhanced arte-
rial phase (b), and subtracted arterial phase (c) sequences demonstrating a right liver lobe treatment
cavity with increased T1 signal intensity on the precontrast sequence and lack of arterial enhance-
ment as evidenced by the subtracted sequence. Baseline increased T1 signal intensity is likely due to
hemorrhagic changes within the lesion following RE. Magnetic resonance T1-weighted gadolinium-
enhanced 20-minute delayed sequence (d) demonstrates a right lobe treatment cavity, decreased
right lobar contrast retention suggesting decreased hepatocyte function and atrophy, and left lobar
contrast retention within normal limits.
126 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

(a)

(b)

Figure 6.3 Volumetric rendering using Vitrea software (Vital Imaging, Inc., Minnetonka, Minnesota) of
the left lobe at the time of diagnosis (a) and 3 months following 90Y radioembolization lobectomy (b)
demonstrating interval hypertrophy of the future liver remnant.

hypertrophy were noted. At 3-month follow- right lobe hepatectomy, the patient remains free
up, complete mRECIST response was noted in of recurrence.
the right lobe HCC (Figure 6.2). Left liver lobe
volumes prior to and 3 months following radio- 6.7.2 SAMPLE RADIATION
embolization lobectomy are provided (Figure SEGMENTECTOMY CASE
6.3). The patient underwent surgical right lobe
hepatectomy 5 months following radioemboli- A 75-year-old male with a past medical history
zation lobectomy. As such, this case represents significant for nonalcoholic hepatic steatosis
successful treatment of the right hepatic tumor was screened for liver cancer using contrast-
along with left lobe hypertrophy in preparation enhanced magnetic resonance imaging revealing
for surgical right lobectomy. After three years of a right hepatic dome HCC (Figure 6.4). While
6.7 Clinical case examples / 6.7.2 Sample radiation segmentectomy case 127

(a) (b)

Figure 6.4 Magnetic resonance T1-weighted gadolinium-enhanced arterial phase (a) and portal
venous phase (b) sequences demonstrating a hepatic dome mass measuring 2.5 cm with early arte-
rial enhancement and portal venous washout consistent with hepatocellular carcinoma. Note the
location of the lesion, which is adjacent to the inferior vena cava, the right hepatic vein, and the
diaphragm.

the size of the lesion (2.5 cm in greatest axial Given the segmental administration of the dose
diameter) is amenable to percutaneous ablation, calculated for lobar infusion, we can calculate the
the location of the lesion adjacent to the inferior dose delivered using Equation 6.1 with a 5% lung
vena cava, the right hepatic vein, and the dia- shunt fraction, minimal dose remaining within
phragm limit the role of percutaneous ablation. the vial (1%) at completion of radioembolization,
The patient was listed for liver transplantation, and a segmental volume of 89.1 mL (0.09 kg utiliz-
received sorafenib therapy, and scheduled for ing the 1.05 kg/L conversion):
radioembolization segmentectomy following a
tumor board conference consisting of transplant Ao (GBq ) (1 − SF )(1 − R ) ⋅ 49.98 (J ⋅ s)
Ddelivered (Gy ) = 
surgeons, hepatologists, medical oncologists, M liver (kg)
and interventional radiologists. The volume
of the right hepatic lobe was found to be 789.9 2 GBq ⋅ (1 − 0.05) ⋅ (1 − 0.01) ⋅ 49.98 (J ⋅ s)
Ddelivered (Gy ) = 
mL; utilizing the conversion factor of 1.05 kg/L, 0.09 Kg
the mass of the right lobe was calculated at 0.83
2 GBq ⋅ 0.95 ⋅ 0.99 ⋅ 49.98 (J ⋅ s)
kg. The activity calculations utilizing the MIRD Ddelivered (Gy ) = 
model described in Chapter 5 are provided—spe- 0.09 Kg
cifically utilizing Equation 5.9: Ddelivered = 1042 Gy

At follow-up imaging, the patient demon-


Davg  (Gy ) ⋅ M liver (kg) strated mRECIST complete response with pro-
Ao  (GBq ) = gressive decrease in size of both the treatment
49.98 (J ⋅ s)
cavity and the treated segment (Figure 6.5).
 120  Gy ⋅ 0.83kg The patient has remained tumor free with con-
Ao  (GBq ) =
49.98 (J ⋅ s) tinued HCC surveillance 3 years after radiation
segmentectomy.
Ao = 2.00  GBq
128 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

(a) (b)

(c)

Figure 6.5 Magnetic resonance T1-weighted gadolinium-enhanced arterial phase (a) at the time of
diagnosis. Magnetic resonance T1-weighted gadolinium-enhanced 20-minute delayed sequences
3 months after radioembolization (b) and 2 years after radioembolization (c) demonstrate progres-
sive decrease in size of both the treatment cavity and the treated segment, which demonstrates
decreased contrast retention suggesting decreased hepatocyte function and atrophy.

REFERENCES
6.8 CONCLUSIONS
Ahmadzadehfar, H., Meyer, C., Ezziddin, S. et al.
The preceding discussion and sample cases (2013). Hepatic volume changes induced
highlight the range of radioembolization appli- by radioembolization with 90Y resin micro-
cations available to physicians for patients with spheres. A single-centre study. Eur J Nucl
varying disease severity. From BCLC A patients Med Mol Imaging 40:80–90.
with anatomically challenging lesions otherwise Aoki, T., Imamura, H., Hasegawa, K. et al.
amenable to ablation to patients with meta- (2004). Sequential preoperative arterial and
static disease deemed unresectable at the time portal venous embolizations in patients
of diagnosis, radioembolization offers solutions with hepatocellular carcinoma. Arch Surg
in a variety of challenging clinical situations. 139:766–774.
Understanding the underlying radiation and Atassi, B., Bangash, A.K., Lewandowski,
tumor biology, dosimetry considerations, and R.J. et al. (2008). Biliary sequelae follow-
patient selection criteria are integral to proper ing radioembolization with Yttrium-90
application of the breadth of radioembolization microspheres. J Vasc Interven Radiol
techniques. 19:691–697.
References 129

Azoulay, D., Castaing, D., Smail, A. et al. (2000). Gaba, R.C., Lewandowski, R.J., Kulik, L.M. et al.
Resection of nonresectable liver metasta- (2009). Radiation lobectomy: preliminary
ses from colorectal cancer after percuta- findings of hepatic volumetric response to
neous portal vein embolization. Ann Surg lobar yttrium-90 radioembolization. Ann Surg
231:480–486. Oncol 16:1587–1596.
Ben-Josef, E., Normolle, D., Ensminger, W.D. Garin, E., Lenoir, L., Rolland, Y. et al. (2012).
et al. (2005). Phase II trial of high-dose confor- Dosimetry based on 99mTc-macroaggregated
mal radiation therapy with concurrent hepatic albumin SPECT/CT accurately predicts tumor
artery floxuridine for unresectable intrahe- response and survival in hepatocellular carci-
patic malignancies. J Clin Oncol Official J Am noma patients treated with 90Y-loaded glass
Soc Clin Oncol 23:8739–8747. microspheres: Preliminary results. J Nucl Med
Braat, A.J., Smits, M.L., Braat, M.N. et al. (2015). Official Publ Soc Nucl Med 53:255–263.
90Y Hepatic radioembolization: An update Garlipp, B., de Baere, T., Damm, R. et al. (2014).
on current practice and recent develop- Left-liver hypertrophy after therapeutic
ments. J Nucl Med Official Publ Soc Nucl Med right-liver radioembolization is substantial
56:1079–1087. but less than after portal vein embolization.
Campbell, A.M., Bailey, I.H., Burton, M.A. (2000). Hepatology 59:1864–1873.
Analysis of the distribution of intra-arterial Ho, S., Lau, W.Y., Leung, T.W. et al. (1996).
microspheres in human liver following hepatic Partition model for estimating radiation doses
yttrium-90 microsphere therapy. Phys Med from yttrium-90 microspheres in treating
Biol 45:1023–1033. hepatic tumours. Eur J Nucl Med 23:947–952.
Chen, M.H., Wei, Y., Yan, K. et al. (2006). Inarrairaegui, M., Pardo, F., Bilbao, J.I. et al.
Treatment strategy to optimize radiofre- (2012). Response to radioembolization with
quency ablation for liver malignancies. J Vasc yttrium-90 resin microspheres may allow
Interven Radiol 17:671–683. surgical treatment with curative intent and
Clement, B., Grimaud, J.A., Campion, J.P. et al. prolonged survival in previously unresectable
(1986). Cell types involved in collagen and hepatocellular carcinoma. Eur J Surg Oncol
fibronectin production in normal and fibrotic J Eur Soc Surg Oncol Br Assoc Surg Oncol
human liver. Hepatology 6:225–234. 38:594–601.
Edeline, J., Lenoir, L., Boudjema, K. et al. (2013). Jakobs, T.F., Saleem, S., Atassi, B. et al. (2008).
Volumetric changes after (90)y radioemboliza- Fibrosis, portal hypertension, and hepatic
tion for hepatocellular carcinoma in cirrho- volume changes induced by intra-arterial
sis: An option to portal vein embolization radiotherapy with 90yttrium microspheres.
in a preoperative setting? Ann Surg Oncol Dig Dis Sci 53:2556–2563.
20:2518–2525. Kim, M.J., Jang, J.W., Oh, B.S. et al. (2013).
Fajardo, L.F., Colby, T.V. (1980). Pathogenesis of Change in inflammatory cytokine profiles
veno-occlusive liver disease after radiation. after transarterial chemotherapy in patients
Arch Pathol Lab Med 104:584–588. with hepatocellular carcinoma. Cytokine
Fernandez-Ros, N., Inarrairaegui, M., Paramo, 64:516–522.
J.A. et al. (2015). Radioembolization of hepa- Kubota, K., Makuuchi, M., Kusaka, K. et al. (1997).
tocellular carcinoma activates liver regenera- Measurement of liver volume and hepatic
tion, induces inflammation and endothelial functional reserve as a guide to decision-mak-
stress and activates coagulation. Liver Int ing in resectional surgery for hepatic tumors.
Official J Int Assoc Study Liver 35:1590–1596. Hepatology 26:1176–1181.
Fernandez-Ros, N., Silva, N., Bilbao, J.I. et al. Lau, W.Y., Leung, T.W., Ho, S. et al. (1994).
(2014). Partial liver volume radioembolization Diagnostic pharmaco-scintigraphy with
induces hypertrophy in the spared hemiliver hepatic intra-arterial technetium-99m macro-
and no major signs of portal hypertension. aggregated albumin in the determination of
Official J Int Hepato Pancreato Biliary Assoc tumour to non-tumour uptake ratio in hepato-
16:243–249. cellular carcinoma. Br J Radiol 67:136–139.
130 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

Lawrence, T.S., Robertson, J.M., Anscher, M.S. to major hepatectomy for colorectal metasta-
et al. (1995). Hepatic toxicity resulting from ses. Ann Surg Oncol 16:1202–1207.
cancer treatment. Int J Radiat Oncol Biol Phys Pompili, M., Saviano, A., de Matthaeis, N. et al.
31:1237–1248. (2013). Long-term effectiveness of resec-
Lencioni, R., Della Pina, C., Bartolozzi, C. (2005). tion and radiofrequency ablation for single
Percutaneous image-guided radiofrequency hepatocellular carcinoma </=3 cm. Results
ablation in the therapeutic management of of a multicenter Italian survey. J Hepatol
hepatocellular carcinoma. Abdominal Imaging 59:89–97.
30:401–408. Rhee, T.K., Omary, R.A., Gates, V. et al. (2005).
Lewandowski, R.J., Kulik, L.M., Riaz, A. et al. The effect of catheter-directed CT angiogra-
(2009). A comparative analysis of trans- phy on yttrium-90 radioembolization treat-
arterial downstaging for hepatocellular ment of hepatocellular carcinoma. J Vasc
carcinoma: Chemoembolization versus Interven Radiol 16:1085–1091.
radioembolization. Am J Transpl Official Riaz, A., Gates, V.L., Atassi, B. et al. (2011).
J Am Soc Transpl Am Soc Transpl Surg Radiation segmentectomy: A novel approach
9:1920–1928. to increase safety and efficacy of radio-
Livraghi, T., Meloni, F., Di Stasi, M. et al. (2008). embolization. Int J Radiat Oncol Biol Phys
Sustained complete response and complica- 79:163–171.
tions rates after radiofrequency ablation of Riaz, A., Kulik, L., Lewandowski, R.J. et al.
very early hepatocellular carcinoma in cirrho- (2009b). Radiologic-pathologic correlation of
sis: Is resection still the treatment of choice? hepatocellular carcinoma treated with internal
Hepatology 47:82–89. radiation using yttrium-90 microspheres.
Llovet, J.M., Bru, C., Bruix, J. (1999). Prognosis Hepatology 49:1185–1193.
of hepatocellular carcinoma: The BCLC Riaz, A., Lewandowski, R.J., Kulik, L.M. et al.
staging classification. Semin Liver Dis (2009a). Complications following radioembo-
19:329–338. lization with yttrium-90 microspheres: A com-
Llovet, J.M., Ricci, S., Mazzaferro, V., Hilgard, P. prehensive literature review. J Vasc Interven
(2008). Sorafenib in advanced hepatocellular Radiol 20:1121–1130; quiz 1131.
carcinoma. N Engl J Med 359:378–390. Salem, R., Mazzaferro, V., Sangro, B. (2013).
Michalopoulos, G.K., DeFrances, M.C. (1997). Yttrium 90 radioembolization for the treat-
Liver regeneration. Science 276:60–66. ment of hepatocellular carcinoma: Biological
Murthy, R., Nunez, R., Szklaruk, J. et al. (2005). lessons, current challenges, and clinical per-
Yttrium-90 microsphere therapy for hepatic spectives. Hepatology 58:2188–2197.
malignancy: Devices, indications, technical Salem, R., Thurston, K.G. (2006a).
considerations, and potential complications. Radioembolization with yttrium-90 micro-
Radiogr A Rev Publ Radiol Soc N Am 25 spheres: a state-of-the-art brachytherapy
(Suppl 1):S41–S55. treatment for primary and secondary liver
Ogata, S. Belghiti, J., Farges, O. et al. (2006). malignancies: Part 3: Comprehensive lit-
Sequential arterial and portal vein emboliza- erature review and future direction. J Vasc
tions before right hepatectomy in patients Interven Radiol 17:1571–1593.
with cirrhosis and hepatocellular carcinoma. Salem, R., Thurston, K.G. (2006b).
Br J Surg 93:1091–1098. Radioembolization with 90yttrium micro-
Padia, S.A., Kwan, S.W., Roudsari, B. et al. (2014). spheres: a state-of-the-art brachytherapy
Superselective yttrium-90 radioemboliza- treatment for primary and secondary liver
tion for hepatocellular carcinoma yields high malignancies. Part 2: Special topics. J Vasc
response rates with minimal toxicity. J Vasc Interven Radiol 17:1425–1439.
Interven Radiol 25:1067–1073. Salem, R., Thurston, K.G. (2006c).
Pamecha, V., Glantzounis, G., Davies, N. et al. Radioembolization with 90Yttrium micro-
(2009). Long-term survival and disease recur- spheres: A state-of-the-art brachytherapy
rence following portal vein embolisation prior treatment for primary and secondary liver
References 131

malignancies. Part 1: Technical and methodo- Teo, J.Y., Allen, J.C., Jr., Ng, D.C. et al. (2015). A
logic considerations. J Vasc Interven Radiol systematic review of contralateral liver lobe
17:1251–1278. hypertrophy after unilobar selective internal
Salem, R., Lewandowski, R.J., Atassi, B. et radiation therapy with Y90. HPB (Oxford).
al. (2005). Treatment of unresectable 2015 Oct 16. doi: 10.1111/hpb.12490 [Epub
hepatocellular carcinoma with use of 90Y ahead of print].
microspheres (TheraSphere): Safety, tumor Teo, J.Y., Goh, B.K., Cheah, F.K. et al. (2014).
response, and survival. J Vasc Interven Underlying liver disease influences volumetric
Radiol 16:1627–1639. changes in the spared hemiliver after selec-
Salem, R., Lewandowski, R.J., Mulcahy, M.F. et al. tive internal radiation therapy with 90Y in
(2010). Radioembolization for hepatocellular patients with hepatocellular carcinoma. J Dig
carcinoma using Yttrium-90 microspheres: A Dis 15:444–450.
comprehensive report of long-term out- Theysohn, J.M., Ertle, J., Muller, S. et al.
comes. Gastroenterology 138:52–64. (2014). Hepatic volume changes after lobar
Salem, R., Vouche, M., Habib, A. et al. (2015). selective internal radiation therapy (SIRT)
Reply: to PMID 24691943. Hepatology of hepatocellular carcinoma. Clin Radiol
61:407. 69:172–178.
Sangro, B., Bilbao, J.I., Boan, J. et al. (2006). Uliel, L., Royal, H.D., Darcy, M.D. et al. (2012).
Radioembolization using 90Y-resin micro- From the angio suite to the gamma-camera:
spheres for patients with advanced hepato- Vascular mapping and 99mTc-MAA hepatic
cellular carcinoma. Int J Radiat Oncol Biol perfusion imaging before liver radioembo-
Phys 66:792–800. lization—A comprehensive pictorial review.
Sangro, B., Gil-Alzugaray, B., Rodriguez, J Nucl Med Official Publ Soc Nucl Med
J. et al. (2008). Liver disease induced 53:1736–1747.
by radioembolization of liver tumors: Vouche, M., Habib, A., Ward, T.J. et al. (2014).
Description and possible risk factors. Cancer Unresectable solitary hepatocellular car-
112:1538–1546. cinoma not amenable to radiofrequency
Sangro, B., Salem, R., Kennedy, A. et al. (2011). ablation: Multicenter radiology-pathology
Radioembolization for hepatocellular carci- correlation and survival of radiation segmen-
noma: A review of the evidence and treat- tectomy. Hepatology 60:192–201.
ment recommendations. Am J Clin Oncol Vouche, M., Lewandowski, R.J., Atassi,
34:422–431. R. et al. (2013). Radiation lobectomy:
Sarfaraz, M., Kennedy, A.S., Cao, Z.J. et al. Time-dependent analysis of future liver
(2003). Physical aspects of yttrium-90 micro- remnant volume in unresectable liver
sphere therapy for nonresectable hepatic cancer as a bridge to resection. J Hepatol
tumors. Med Phys 30:199–203. 59:1029–1036.
Seror, O., Nault, J.C., Nahon, P. et al. (2015). Yamazaki, H., Oi, H., Matsumoto, K. et al. (1996).
Is segmental transarterial yttrium 90 Biphasic changes in serum hepatocyte growth
radiation a curative option for solitary hepa- factor after transarterial chemoemboliza-
tocellular carcinoma ≤5 cm? Hepatology tion therapy for hepato-cellular carcinoma.
61:406–407. Cytokine 8:178–182.
Shindoh, J., Tzeng, C.W., Aloia, T.A. et al. (2013). Yip, D., Allen, R., Ashton, C., Jain, S. (2004).
Optimal future liver remnant in patients Radiation-induced ulceration of the stomach
treated with extensive preoperative chemo- secondary to hepatic embolization with radio-
therapy for colorectal liver metastases. Ann active yttrium microspheres in the treatment
Surg Oncol 20:2493–2500. of metastatic colon cancer. J Gastroenterol
Siddiqi, N.H., Devlin, P.M. (2009). Radiation Hepatol 19:347–349.
lobectomy—A minimally invasive treatment Yoo, H., Kim, J.H., Ko, G.Y. et al. (2011).
model for liver cancer: Case report. J Vasc Sequential transcatheter arterial chemo-
Interven Radiol 20:664–669. embolization and portal vein embolization
132 Radioembolization in segmentectomy, lobectomy, and future liver remnant hypertrophy

versus portal vein embolization only before Zorzi, D., Laurent, A., Pawlik, T.M. et al. (2007).
major hepatectomy for patients with Chemotherapy-associated hepatotoxicity and
hepatocellular carcinoma. Ann Surg Oncol surgery for colorectal liver metastases. Br J
18:1251–1257. Surg 94:274–286.
PART 3
Treating Patients with
Radioembolization

7 Radiation safety concerns associated with preparing the dosage, treating


and releasing the patient, and managing radioactive waste 135
William D. Erwin
8 The radiation biology of radioembolization 167
Marta Cremonesi, Francesca Botta, Mahila Ferrari, Lidia Strigari, Guido Bonomo,
Franco Orsi, and Roberto Orecchia
9 Microsphere deposition, dosimetry, radiobiology at the cell-scale,
and predicted hepatic toxicity 199
Stephan Walrand
7
Radiation safety concerns associated
with preparing the dosage, treating
and releasing the patient, and
managing radioactive waste

WILLIAM D. ERWIN

7.1 Introduction 135 7.4 Radiation safety during treatment in


7.2 The written directive 136 the IR suite 151
7.2.1 Authorized user 137 7.4.1 Immediate preadministration 151
7.2.2 Yttrium-90 microspheres 7.4.2 During administration 153
absorbed dose-related 7.4.3 Immediate postadministration 154
properties 138 7.5 Patient release 155
7.2.3 SIR-Spheres prescription 138 7.5.1 To the public 156
7.2.4 TheraSphere prescription 140 7.5.2 Hospital admission 157
7.2.5 Alternative radioembolization 7.6 Significant posttreatment events 158
prescription methods 140 7.6.1 Surgery: Liver resection or
7.2.6 Reportable medical event 141 transplant, or other 158
7.3 Dosage delivery to and preparation in 7.6.2 Autopsy, burial, or cremation 158
the hot lab 142 7.7 Radioactive waste 159
7.3.1 SIR-Spheres dosage dispensing 7.7.1 Residual activity assay 159
and preparation 144 7.7.2 Example procedure
7.3.2 TheraSphere dosage measurements and calculations 160
dispensing and preparation 145 7.7.3 Storage for decay and disposal 162
7.3.3 Exposure rate measurement of 7.8 Conclusions 163
dosage vials 148 References 163
7.3.4 Dosage cart preparation 148

radiation safety concerns. Among these are patient


7.1 INtrODUCtION absorbed dose prescription or radioactivity pre-
scription (also referred to as dosage throughout
Radioembolization, being a form of internal this chapter) and associated calculations; radioac-
administration of radioactive material (RAM) in tivity (heretofore abbreviated to activity) delivery
humans, necessarily has associated with it a num- to and preparation in the radiopharmacy (or hot
ber of patient, personnel, and general public-related lab); transport to interventional radiology (IR) and

135
136 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

infusion into the patient; handling of the radio- States as permanent implant manual brachy-
active waste and contamination from the proce- therapy sources, as approval for both radioem-
dure; internal radiation exposure of the patient; bolization products as a device rather than a
and external exposure from both the patient and radiopharmaceutical was sought and obtained
the radioactive waste (Salem and Thurston, 2006; from the FDA. However, the NRC has developed
Gulec and Siegel, 2007; Dezarn et al., 2011). These a Microsphere Brachytherapy Sources and Devices
various radiation safety aspects of radioemboliza- licensing guidance document (NRC, 2012) relat-
tion are the subject of this chapter. ing how its “Medical Use of Byproduct Material”
The two radiolabeled microspheres products (NRC, 2013) regulations apply to radioemboliza-
approved by the Food and Drug Administration tion due to its unique nature (the infusion of mil-
(FDA) for liver-directed radioembolization, lions of individual sources in solution directly
TheraSphere® (BTG International Ltd., Canada) into the hepatic arterial bloodstream). The NRC
and SIR-Spheres® (Sirtex Medical Limited, Lane has chosen to categorize radioembolization under
Cove, NSW, Australia), have both similarities 10 (Code of Federal Regulations CFR) 35 Subpart
and differences from the perspective of radiation K—Other Medical Uses of Byproduct Material
safety (Dezarn et al., 2011). On the one hand, both or Radiation From Byproduct Material (10 CFR
employ the same radionuclide for the β radiother- 35.1000) as opposed to Subpart F—Manual
apy (yttrium-90, 90Y), as well as a similar range of Brachytherapy; and the guidelines describe how
particle sizes (20–60 μm and 20–30 μm spheres, licensees comply with certain aspects of the 10
respectively). The radiopharmaceutical 99mTc- CFR 35 general requirement Subparts A, B, C, L,
radiolabeled macroaggregated albumin (MAA) is and M. The guidelines specifically address autho-
employed for both as a surrogate for pretreatment rized user (AU) training and experience; the writ-
microcatheter placement imaging, lung shunt ten directive; 90Y microspheres source leak tests
quantification, and treatment planning absorbed (explicitly stated as not applicable) and inventory;
dose or activity calculations. Finally, the admin- patient release; labeling; medical event reporting;
istration kits of both have the same basic compo- and waste disposal issues related to radionuclidic
nents: a sealed v-vial containing the microspheres impurities in the microspheres. Certain important
housed in an acrylic β shield; an acrylic infusion statements in the NRC guidelines regarding the
delivery box in which the v-vial-in-shield is placed various radiation safety aspects of radioemboliza-
that serves as additional β radiation shielding as tion covered in this chapter will be mentioned in
well as a RAM containment vessel; and needles the corresponding sections that follow.
and associated infusion lines (tubing) for connec-
tion to and flow between the injection syringe,
v-vial, and the prepositioned microcatheter. On
the other hand, TheraSphere consists of 3.29 g/ 7.2 tHE WrIttEN DIrECtIVE
mL specific gravity glass spheres with naturally
occurring 89Y embedded in the material that are Internal administration of a therapeutic amount of
irradiated in a nuclear reactor to produce 90Y via RAM requires, by regulation, what is called a writ-
the 89Y(n,γ) 90Y reaction (with a number of radio- ten directive, defined by the NRC as “an authorized
nuclidic impurities being produced in the process), user’s (AU) written order for the administration of
while SIR-Spheres consists of lower density (1.6 g/ byproduct material or radiation from byproduct
mL) resin-based spheres with 90Y extracted from material to a specific patient or a human research
a strontium-90 (90Sr)/90Y generator and chemically subject” (NRC, 2013). The information required to
bound to the surface, which results in no radionu- be present in a written directive is specific to the
clidic impurities other than a trace amount of 90Sr category under which a particular RAM therapy
due to “breakthrough” in the extraction process. falls, and there are currently six categories defined
Finally, a higher amount of activity, in general, is in the NRC regulations. Radioembolization, due to
administered with TheraSphere. its unique nature, has characteristics that resem-
Yttrium-90 microspheres are recognized in ble those in both “unsealed byproduct material
the United States by the Nuclear Regulatory other than sodium iodide I-131” and “all other
Commission (NRC) and associated Agreement brachytherapy” categories. As a consequence, the
7.2 The written directive / 7.2.1 Authorized user 137

NRC has established special case written directive Radiation Oncologist). The NRC recognizes three
requirements for 90Y microspheres liver-directed types of AU for 90Y microspheres liver-directed
therapy (NRC, 2012). First, the prescription can therapy (NRC, 2012):
be in terms of either radiation absorbed dose (rad
or Gy) or activity (mCi or GBq). The documented 1. A physician board certified in radiation
written directive must include (in addition to the oncology, qualified and identified as an AU
obvious patient identification, date, and AU signa- on a licensee’s RAM license for manual
ture) the following information: brachytherapy
Prior to treatment: 2. A physician board certified in nuclear medi-
cine, qualified and identified as an AU on a
1. Treatment site (anatomical target, e.g., whole licensee’s RAM license for all therapeutic uses
liver) of unsealed RAM
2. Radionuclide and physical form (i.e., “90Y 3. An IR physician meeting a specific set of
microspheres”) requirements related to 90Y microsphere’s liver-
3. Prescribed absorbed dose or activity directed therapy:
4. Manufacturer of the particular 90Y micro- a. 80 hours of didactic and laboratory train-
spheres device used ing in the radiation physics, instrumenta-
5. The statement “or dose delivered at stasis” or tion, protection, activity measurement
“or activity delivered at stasis” (if appropriate, (assay), and biology aspects of RAM
e.g., for SIR-Spheres) (including 90Y)
6. Maximum acceptable absorbed dose or activity b. 90Y microspheres AU supervised work
in the lungs and any other possible sites (e.g., experience:
gastrointestinal tract) due to shunting i. RAM ordering, receiving, surveying,
and unpacking
After treatment (within 24 hours): ii. Activity assay instrument quality con-
trol procedures
1. Absorbed dose or activity delivered to the iii. Absorbed dose and activity aspects of
treatment site radioembolization treatment planning
2. Absorbed dose or activity delivered to nontar- iv. 90Y microspheres activity calculation,
get sites (e.g., lungs) assay, and preparation
3. If either stasis or a patient condition emerging v. Medical event prevention administra-
during the treatment occurred and prevented the tive controls
full absorbed dose or activity from being deliv- vi. RAM spill containment and decon-
ered (≥80% of prescribed, per NRC regulations), tamination procedures, including those
a statement to that effect must be recorded specific to microspheres
vii. Radioembolization patient case history
(If multiple sites, e.g., right and left lobes of the follow-up and review
liver, are treated separately, then a separate written
directive is required for each.) In addition to the aforementioned require-
ments, each candidate AU must undergo docu-
7.2.1 AUTHORIZED USER mented training on the operation, safety, and
clinical applications of each 90Y microspheres
AU status for the medical use of RAMs is restricted delivery system for which AU status is sought and
to those physicians (or dentists or podiatrists) successfully complete at least three hands-on treat-
who meet certain minimum didactic, labora- ments under the direct supervision of either the
tory, and clinical training, as well as board certi- manufacturer or a current AU of the delivery sys-
fication and licensure requirements (NRC, 2015). tem. In the case of training by the manufacturer,
These requirements are specific to each diag- at least three directly supervised in vitro simulated
nostic and therapeutic use category and corre- treatments should be performed first. Finally, the
spond to specific physician specialties (e.g., “AU AU is responsible for ensuring that all person-
Eligible” Radiologist, Nuclear Medicine Physician, nel to whom the AU delegates 90Y microsphere
138 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

preparation, measurement, absorbed dose calcula- secondary bremsstrahlung x-ray and γ radiations
tions, and administration are adequately trained do occur. However, their yields are quite small,
(e.g., AU ineligible interventional radiologists to and thus 100% local deposition is a reasonable
whom the AU has delegated performing the actual approximation.)
infusion of the microspheres). The mean energy emitted per 90Y decay is
1.498E–13 Gy-kg/Bq-s (Eckerman and Endo, 2008);
and the two microspheres products are biocom-
7.2.2 YTTRIUM-90 MICROSPHERES patible but not biodegradable, and therefore are
ABSORBED DOSE-RELATED permanent implants once they are trapped at the
PROPERTIES capillary level (in either tumors, the normal liver
or the lungs). Thus, no 90Y is cleared biologically,
Before presenting the specifics of the methods and all activity decays in place with an effective
employed for determining the absorbed dose or half-life equal to the physical half-life of 90Y (64.24
activity required for SIR-Spheres and TheraSphere hours). The cumulative energy deposited in tissue
radioembolization, it is important to understand per GBq of 90Y microspheres in that tissue is
the underlying radiation absorbed dose-related
properties of 90Y microspheres upon which the pre- 1.498E − 13 Gy ⋅ kg / Bq ⋅ s
scriptions of the two products are currently based. × 1E9 Bq / GBq
Yttrium-90 is considered a pure beta (β) emitter, × 3.6E3 s / h × 64.24 h
49.98 Gy ⋅ kg / GBq = (7.1)
whereby (1) 100% of its decay occurs via β– emis- ln(2)
sion to a stable daughter as opposed to a β-emitting
radionuclide that also has significant alternative where 64.24 hours/ln(2) is the integral of e–ln(2)t/64.24 h
electron (e–) capture and/or positron (e+) branch- from t = 0 to infinity (Dezarn et al., 2011) (49.98 is
ing fractions (e.g., copper-64). (2) Its secondary often rounded off to 50). This greatly simplifies the
Auger and internal conversion (IC) e–, gamma calculation of absorbed dose at the level of tumors,
(γ), and characteristic daughter x-rays emissions normal liver, and lungs and is exploited by the
are negligible (Eckerman and Endo, 2008; Dezarn current methods of prescribing SIR-Spheres and
et al., 2011). The dominant (99.9885%) β– emission TheraSphere.
has average/endpoint energies of 0.935 MeV/2.280
MeV, decaying to ground state zirconium-90 (90Zr).
The remaining decays (0.0115%) are lower-energy 7.2.3 SIR-SPHERES PRESCRIPTION
(0.186 MeV/0.519 MeV) β– emissions resulting in a
1.760 MeV excited state of 90Zr, which immediately Three methods of prescribing SIR-Spheres have
transitions to its ground state via either IC (domi- been developed and recommended by the manu-
nant), a two γ-emission or e+–e– pair production facturer; two are activity-based empiric (so-called
(Ford, 1955; Johnson, 1955; Selwyn et al., 2007). (A “basic” and “body surface area” or “BSA”) meth-
third, even lower-energy emission occurs, but its ods, and the third is an absorbed dose-based par-
yield is so small that it can be ignored.) As a con- tition model (Sirtex Medical Limited, 2003, 2010;
sequence, essentially all of the 90Y decay energy Salem and Thurston, 2006; Dezarn et al., 2011;
emitted (excluding that carried away by antineu- Lam et al., 2014; Braat et al., 2015). All three will
trinos) is e– kinetic energy. The ranges in tissue be described, although the basic empiric method
for the average (0.935 MeV) and maximum (2.280 is now considered a legacy method, having been
MeV) energy electrons emitted by 90Y are 4.0 and essentially superseded by the BSA method.
11.3 mm, respectively (Cole, 1969; ICRU, 1984); SIR-Spheres was originally developed as a
and 50% and 90% of the absorbed dose from a whole-liver treatment for hepatic metastases from
point source of 90Y are deposited within 2.3 and colorectal cancer with adjuvant chemotherapy,
5.2 mm, respectively (Berger, 1976). Thus, 100% of both delivered via the hepatic artery; and the 3
the energy emitted can be assumed to be depos- GBq maximum administered activity and 0.20
ited locally at a macroscopic level of an organ or lung shunt fraction (SF) limits were established
tumor (Pasciak and Erwin, 2009; Dezarn et al., empirically early on (Kennedy et al., 2007). These
2011). (Some energy is not absorbed locally, as limits correspond to maximum tolerated liver and
7.2 The written directive / 7.2.3 SIR-spheres prescription 139

lung absorbed doses of 80 and 30 Gy, respectively. supplanted the basic method as the empiric method
The 3 GBq limit was derived from a worst-case of choice. (The reduction in the base amount of
uniform whole-liver uptake and 80 Gy absorbed activity to administer as a function of % SF is iden-
dose with no lung shunting, the energy deposited tical to that used for the basic method.)
per GBq, and a 1.91 kg reference adult liver mass: The basic formula for the BSA method
(Equation 5.7) assumes a whole-liver treatment.
80  Gy × 1.91 kg However, lobar (either separate left and right or
3 GBq = (7.2)
50 Gy ⋅ kg / GBq just one or the other) and segmental SIR-Spheres
treatments have become commonplace over time.
The maximum SF (0.20) was derived from a 30 The total activity for lobar or segmental treatments
Gy lung absorbed dose limit, the energy deposited is typically scaled according to their estimated
per GBq, a 1 kg reference adult male lung mass, percentage of total liver volume (Equation 5.8).
and the maximum 3 GBq: The standard (nominal normal) right/left lobar
volume split is 70%/30%, but the split is typically
30 Gy × 1 kg determined on a patient-by-patient basis as dis-
0.20 = (7.3)
50 Gy ⋅ kg / GBq × 3 GBq eased liver anatomy often diverges from normal
and % TI between lobes and segments to be treated
is often variable.
7.2.3.1 Empiric methods The pretreatment part of the written directive
The basic empiric method starts with the 3 GBq for the BSA method should include the lung shunt
activity limit and scales the activity downward to a (% or fraction), the prescribed activity for each
more conservative level according to both the esti- treatment target anatomy (whole liver, lobe, or seg-
mated fractional tumor involvement (TI) in the ment) A(GBq) (Equation 5.8), and the activity to
liver and SF as shown in Tables 7.1 and 7.2. be delivered to the lungs for each treatment target:
The BSA method Chapter 5, Section 5.8.2 was
later developed after unacceptable levels of clinical A (GBq) × SF (7.4)
and laboratory toxicities were found when using
the basic method. The BSA formula for calcula- as well as the overall cumulative activity to be
tion of the base amount of activity to administer delivered to the lungs for all treatments. The post-
(Equations 5.6 and 5.7) was developed empirically; treatment part should include the net total and
and although the basis upon which the specific for- lung activities delivered for each treatment:
mula parameters were derived is nowhere reported
in the literature, it has nonetheless effectively A (GBq) × (1 − W ) (7.5)

Table 7.1 SIR-Spheres basic empiric method A (GBq) × SF × (1 − W ) (7.6)


base prescribed activity level
as well as net overall cumulative activity delivered
% tumor GBq to the lungs. Here, A(GBq) is the preinfusion dose
>50 3 calibrator assay of the treatment target prescribed
25≤% tumor≤50 2.5 activity (see Section 7.3); W is the infusion waste
<25 2 activity fraction computed as the ratio of the infu-
sion waste activity and pretreatment A(GBq) sur-
vey meter readings (see Section 7.3 and 7.7), and
Table 7.2 SIR-Spheres basic and BSA empiric
all measurements are decay corrected to the time
method prescribed activity level reduction
of infusion.
% Lung shunt GBq % reduction
<10 0 7.2.3.2 Partition model method
10 ≤ % LS < 15 20
The third manufacturer-recommended method of
15 ≤ % LS ≤ 20 40
prescribing SIR-Spheres is a more robust, patient-
>20 No treatment
specific, and absorbed dose-based method called
140 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

the “partition model” (Ho et al., 1996; Sirtex lung shunt (% or fraction), the tumor, normal liver,
Medical Limited, 2003; Dezarn et al., 2011; Braat and lung masses and prescribed absorbed doses,
et al., 2015), whereby the total administered activ- and prescribed total activity A0(GBq). The post-
ity A0(GBq) is assumed to be apportioned among treatment part should include the net total activity
three partitions—tumor, normal liver, and lung— infused and adjusted tumor, normal liver, and lung
according to relative blood flow and SF Chapter absorbed doses.
5, Section 5.8.4). The activity to administer is
back calculated from prescribed target tumor 7.2.4 THERASPHERE PRESCRIPTION
(Dtumor(Gy)), normal liver (Dnormal(Gy)), and lung
(Dlung(Gy)) maximum tolerated absorbed doses The current manufacturer-recommended method
and estimates of tumor-to-normal liver blood of prescribing TheraSphere consists of calculating
flow ratio (T:N), target tumor (Mtumor(kg)), and the infused activity required for the whole liver
normal liver (Mnormal(kg)) masses, and optionally or separate lobar or segmental treatment targets
lung mass, M lung(kg), although 1  kg is typically (A0(GBq)) based on a treatment target absorbed
assumed (the 0.8 kg reference adult female lung dose (80–150 Gy is considered therapeutic) assum-
mass could be used for female patients) as an esti- ing a uniform microsphere distribution (Davg(Gy))
mate of patient-specific lung mass that is difficult and lung 30 Gy (Dlung(Gy)) single treatment session
to obtain. The absorbed dose-based constraints on and 50 Gy cumulative (repeat treatment) absorbed
A0(GBq) are then derived using the model param- dose limits (Chapter 5, Section 5.8.3):
eters (Equations 5.10 through 5.14):
Davg (Gy) × M liver (kg)
Dlung (Gy) × M lung (kg) A0 (GBq) =
Alung (GBq) ≤ (7.7) 49.98 Gy ⋅ kg / GBq × (1 − SF)
(49.98 Gy ⋅ kg / GBq × SF) (7.9)
Dlung (Gy) × M lung (kg)

Dnormal (Gy) × 49.98 Gy ⋅ kg / GBq

( T:N × M tumor (kg)) The 1 kg reference adult male lung mass for
  Mlung (kg) is assumed, unless the reference mass for
+ M 
 normal (kg)  (7.8) female patients (0.8 kg) or a patient-specific lung
Anormal (GBq) ≤
49.98 Gy ⋅ kg / GBq × (1 − SF) mass estimate is employed (Simon, 2000; Salem
and Thurston, 2006; BTG International Ltd., 2010,
The prescribed activity A0(GBq) is constrained 2014; Dezarn et al., 2011; Busse et al., 2013; Braat
by upper limits on Dnormal(Gy) and Dlung(Gy), et al., 2015).
and thus would be constrained to be the lesser The pretreatment part of the written directive
of Anormal(GBq) or A lung(GBq). If A0(GBq) is con- for TheraSphere should include the lung shunt (%
strained, then Dtumor(Gy) and either Dlung(Gy) or or fraction); the mass, prescribed activity, absorbed
Dnormal(Gy) would be recalculated based on the dose for each treatment target and cumulative
constrained value for A0(GBq) and the model dose absorbed dose to the lung. The posttreatment part
equations. The manufacturer recommends lung should include the net total, target, and lung activi-
and normal liver absorbed dose limits of 25 and 80 ties infused and adjusted treatment target and lung
Gy (70 Gy if cirrhotic), respectively (Sirtex Medical absorbed doses (Equation 7.10).
Limited, 2003; Braat et al., 2015), although a 30 Gy
limit for lung and lower values for normal liver, 7.2.5 ALTERNATIVE
e.g., Emami 30 Gy whole-liver external beam RADIOEMBOLIZATION
tolerance dose (TD) 5/5 limit (Emami et al., 1991), PRESCRIPTION METHODS
is commonly employed. The partition model equa-
tions are only valid for solitary tumors or multiple The use of other more sophisticated prescrip-
tumors with identical T:N values, within the treat- tion methods, such as artery-specific partition
ment target. modeling (Kao et al., 2012) or three-dimensional
The pretreatment part of the written directive absorbed dose calculation (absorbed dose point
for the partition model method should include the kernel, Monte Carlo, or deterministic) and
7.2 The written directive / 7.2.6 Reportable medical event 141

treatment-planning techniques analogous to those and what the written notification must contain
used for external beam radiation therapy or brachy- (names of licensee and prescribing physician, brief
therapy (Dieudonné et al., 2011; Petitguillaume description of event and why it occurred, effect
et  al., 2014), is not precluded for radioemboliza- on patient, actions taken or planned to prevent
tion. The accuracy of such methods should be vali- recurrence, and certification that the individual
dated prior to clinical use, and the NRC states in or responsible relative or guardian was notified or
the 90Y microspheres licensing guidance document why not). The conditions applicable to radioembo-
that such alternative methods should be submitted lization for a reportable medical event are (NRC,
(presumably to the NRC or Agreement State regu- 2012):
latory body, although that is not stated explicitly).
1. A dose that differs from the prescribed
absorbed dose or that which would have
7.2.6 REPORTABLE MEDICAL resulted from the prescribed dosage by more
EVENT than
a. 0.05 Sv (5 rem) effective dose equivalent
The NRC requires that the absorbed dose or activ- b. 0.5 Sv (50 rem) equivalent dose to an organ
ity delivered to each treatment target be either or tissue
within 20% or within the therapeutic range of that c. 0.5 Sv (50 rem) shallow dose equivalent to
prescribed, unless the reason for falling outside the skin
those limits was due to intervention by the patient
and
(NRC, 2013). If the infusion was terminated early
due to stasis or other emergent patient condition, it i. The total absorbed dose delivered dif-
is necessary to include a statement to that effect in fers from the prescribed absorbed dose
the posttreatment written directive (NRC, 2012). by >±20%
The ±20% limit of prescribed activity (empiric ii. The total dosage delivered differs from
method) would be applicable to SIR-Spheres. The the prescribed dosage by 20% or more
±20% limit of prescribed absorbed dose would or falls outside the prescribed dosage
be applicable to TheraSphere, SIR-Spheres if the range
partition model was used, and either product if 2. A shallow dose equivalent to the skin, an
an alternative absorbed dose-based method of equivalent dose to an organ or a tissue other
treatment planning was employed. (One could than the treatment site that exceeds
argue that within the prescribed range is applica- a. by 0.5 Sv (50 rem) to an organ or a tissue
ble for TheraSphere, as a delivered absorbed dose b. 50% or more of that expected from the
anywhere between 80 and 150 Gy is considered administration defined in the written
therpeutic [BTG International Ltd., 2014]). If the directive
delivered absorbed dose or activity falls outside
these limits and it was not due to the patient, but (excluding, for permanent implants, seeds that
either a failure of the device, improper technique were implanted in the correct site but migrated
(e.g., use of a microcatheter with too small a bore outside the treatment site).
size, incorrect microcatheter placement or device Calculating the estimated difference between
assembly), or administering the incorrect activity delivered and prescribed absorbed dose for a pos-
(e.g., accidently switching separate left and right sible reportable medical event where either too
lobe treatment dosage vials), then it may have to much or too little activity was inadvertently deliv-
be reported to the NRC or Agreement State regu- ered to the intended target tissue (normal liver,
latory body. The NRC has established conditions lungs, and/or tumor) is relatively straightforward.
under which such a misadministration is con- The target prescribed activity, A(GBq); the target
sidered reportable, as well as timeliness of the mass, M [kg, e.g., computed tomography (CT)
reporting following discovery (notification of the or magnetic resonance (MR) volume based]; the
referring physician and patient within 24 hours, estimated fraction of prescribed activity actu-
phone call within 24 hours, and written report ally delivered to the target, F (can be either less
within 15 days to the NRC or Agreement State), than or greater than 1.0); and the constant 49.98
142 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

Gy-kg/GBq can be combined to compute the esti- [self-attenuation within the source; attenuation by
mated absorbed dose (Gy) actually delivered to the well insert, vial/syringe dipper, or liner (e.g.,
the target tissue: copper) employed; or a combination thereof]. The
external radiation from a source of a given activity
49.98 × A × F (7.10) of a pure β-emitting radionuclide such as 90Y, on
D=
M the other hand, is highly variable. This is due to the
predominance of secondary bremsstrahlung x-ray
and the difference from the prescribed absorbed
radiation, the amount of which is highly dependent
dose then calculated using D.
upon source material composition and geometry,
Estimating the absorbed dose to an unintended
as well as intervening material. As a consequence,
target, whether hepatic or extrahepatic, is more
a separate calibration is needed for each 90Y source
complicated, as a posttherapy 90Y bremsstrahlung
form factor encountered.
single-photon emission computed tomography
Calibration of a dose calibrator for assay of
(SPECT)/CT or positron emission tomography
dosages of either radioembolization product is
(PET)/CT scan from which accurate estimates of
currently accomplished by requiring the manufac-
both absolute activity within and mass of the unin-
turer to ship a certificate of analysis with at least
tended target can be obtained, must be relied upon,
the first three dosages, indicating the calibrated (as
and standardized methods of generating such
opposed to nominal or estimated) activity of the
quantitative scans do not yet exist.
particular dosage with a reasonably small uncer-
tainty, along with the date and time of the activ-
ity calibration (Dezarn et al., 2011). The calibrated
activity of each dosage is decay corrected to the
7.3 DOSaGE DELIVErY tO
time of measurement with each dose calibrator
aND PrEParatION IN providing an expected reading and the dose cali-
tHE HOt LaB brator setting adjusted until the expected reading
is achieved. Repeating this process for at least three
The final form of each radioembolization dosage dosages allows for calculation of an average setting
ready to be administered to the patient is similar in an effort to improve the overall accuracy of sub-
for both products, that is, radiolabeled micro- sequent dosage activity assays. Periodic recalibra-
spheres in solution (sterile water or saline) in an tion (e.g., annually) would be prudent to maintain
infusion v-vial contained within an acrylic shield. equivalence of activity assay with the manufac-
However, the process by which each dosage is dis- turer. (Optimal and consistent placement of the
pensed into that final form currently differs greatly radioembolization dosage within the dose cali-
between the two. Thus, the specifics of dosage brator will yield the best calibration result. A vial/
delivery and preparation of each radioemboliza- syringe dipper such as the Biodex Atomlab model
tion product will be discussed separately. 086-242 allows such placement of both radio-
Licensee verification of the activity of dosages embolization product dosage vials. In addition,
of either radioembolization product requires the the shipping vial containing SIR-Spheres must
calibration of all dose calibrators that will be used first be gently shaken back and forth to suspend
for this purpose. Dose calibrator manufacturers the microspheres uniformly in solution. Tipping
have established standard settings for each of their the vial upside down should be avoided during the
dose calibrators for assaying sources of radionu- process of suspension, as that may cause some of
clides commonly used in nuclear medicine and the microspheres to be trapped around the periph-
that emit an abundance of characteristic gamma ery of the vial’s septum.)
and/or x-ray photons (Carey et al., 2012). For some Calibration of dose calibrators for SIR-
of those radionuclides, minor setting adjustments Spheres dosage assay is currently not traceable
or correction factors based on source form factor to the U.S. National Institutes of Standards and
(e.g., syringe versus vial) may be necessary due to a Technology (NIST), although the Australian
slight variation in the radiation output of abundant Nuclear Science and Technology Organization
low-energy photon emissions and a result of loca- (ANSTO, Australian equivalent of NIST) and the
tion of the source within the dose calibrator well Australian Radiopharmaceuticals and Industrials
7.3 Dosage delivery to and preparation in the hot lab 143

have made activity measurements of SIR-Spheres, with, e.g., that for the 90Y radioimmunoconjugate
and (as of 4 years ago) the manufacturer was still Zevalin®, for which both a NIST-traceable transfer
using an ANSTO-traceable calibrated ion cham- standard (Figure 7.2) and a calibration procedure
ber for activity measurement (Dezarn et al., 2011). exist (Thieme et al., 2004). (One could attempt a self-
The manufacturer of TheraSphere participates in calibration for assaying radioembolization form
the NIST Radioactivity Measurement Assurance factor dosages by first calibrating the dose calibra-
Program, where settings for commercial Capintec tor with the Zevalin transfer standard and assaying
dose calibrators have been established; a secondary a source of 90Y chloride in the Zevalin form factor,
measurement standard for routine calibration is calibrating for microspheres suspended in solu-
maintained; and the manufacturer’s dose calibra- tion in the shipping vial [SIR-Spheres] and v-vial
tor measurement of activity is periodically veri- within its acrylic shield [both products] by dispens-
fied (Dezarn et al., 2011). However, no standards ing a known amount of the calibrated 90Y chloride
organization-traceable dose calibrator standards or activity plus fluid resulting in a volume employed
accredited laboratory calibration methods exist for for the particular radioembolization product, and
either products at this time, although methods for adjusting the dose calibrator setting until the cor-
developing NIST-traceable activity standards and rect reading is obtained. Uncertainties associated
calibrations for SIR-Spheres on an institution-by- with such a calibration include that for the transfer
institution basis have been published (Mo et al., standard itself [95% confidence interval = ±4.7%];
2005; Selwyn et al., 2007, 2008). Until such activ- activity loss during the dispensing that is not
ity standards and methods are available for all accounted for; and the fact that chloride solution
institutions to exploit for periodic dose calibrator does not mimic microspheres exactly, which have
calibration (including SIR-Spheres in variable solu- a density substantially greater than that of water
tion volumes in the shipping vial, as exemplified in and thus for which a larger amount of secondary
Figure 7.1), each institution must rely on cross-cal- bremsstrahlung radiation for the same activity will
ibration with the manufacturer. As a result, there be produced, and which precipitate to the bottom
will most likely be a larger uncertainty in the mea- of the vial as opposed to remaining uniformly sus-
surement of radioembolization dosages compared pended in solution.)

Dose calibrator setting vs. 90Y solution volume in 10mL glass vial
75

70
Calibration number (×10)

65
y = –3.6x + 73.8
y = –4.1x + 74.1
60

55

50

45
0 1 2 3 4 5 6
90Y Solution volume (mL)

Figure 7.1 Settings for two Capintec CRC-15R dose calibrators for assaying 90Y in solution in a 10 mL
glass vial as a function of solution volume. Employing the 5 mL of volume setting to assay 1 mL
of solution results in an overestimation of activity on the order of 10%.
144 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

prescribed SIR-Spheres treatment dosages from


the shipping vial to an infusion v-vial (Figure 7.3b).
Preparation and dispensing of each SIR-Spheres
treatment dosage is a multistep process (Dezarn
et al., 2011). The first step consists of assaying the
initial activity in the shipping vial. The micro-
spheres must be uniformly suspended in solution
(in the same fashion as that for initial dose calibra-
tor calibration), and the vial is assayed immediately
in the dose calibrator using the previously estab-
lished dose calibrator setting. This assay allows
the activity concentration in the shipping vial to
be calculated (by division by 5 cc). The second step
consists of the calculation of the volume to with-
draw for the first dosage by dividing the required
activity (prescribed activity decay-corrected from
the anticipated time of infusion back to the time
of dispensing) by the activity concentration, and
then, transferring that volume from the shipping
Figure 7.2 NIST-traceable 90Sr/90Y transfer vial into the infusion v-vial via a 5 cc syringe. (A 3
standard for calibration of dose calibrators for cc syringe may be preferable for volumes less than
assaying 90Y Zevalin (2σ = 4.7%) that simulates 4 2 cc, for more accurate volume withdrawal.) The
mL of 90Y Zevalin in a 10 mL Becton–Dickinson
third step consists of assaying the activity remain-
plastic syringe. The standard is accompanied by
a table of correction factors for other solution ing in the shipping vial and calculating the activ-
volumes that vary linearly from 0.983 at 9 mL to ity dispensed as the difference between the initial
1.017 at 1 mL. and remaining activities. If the difference is too
high, then too much activity was withdrawn and
some must be transferred back to the shipping vial,
7.3.1 SIR-SPHERES DOSAGE and if too low then additional activity needs to be
DISPENSING AND transferred to the infusion v-vial. (It is worth not-
PREPARATION ing here that dose calibrator cross-calibration with
the manufacturer is only for the initial activity in
SIR-Spheres are shipped from the manufacturer to 5 mL, whereas it can be demonstrated that a 5 mL
the hot lab on the day of treatment, as a nominal volume dose calibrator setting can cause an over-
amount of activity (3 GBq ± 10%) in 5 mL of ster- estimate on the order of 10% in a 1 mL residual
ile water contained within a 10 cc glass vial inside activity assay. On the other hand, no standardized
a lead pot, regardless of dosage (Figure 7.3a). The methodology exists yet for calibrating the dose
activity in the shipped vial is calibrated for 18:00 calibrator for assaying the shipping vial contain-
hours United States Eastern Time (U.S. ET) on ing microspheres in a variable volume of solution.)
the day of treatment, and the product has a shelf- Steps 2 and 3 must be performed quickly, while
life of 24 hours postcalibration (Sirtex Medical the microspheres are kept in suspension, and are
Limited, 2003, 2010; Dezarn et al., 2011). (One may repeated for each additional treatment dosage to
request the delivery of the product for treatment be dispensed. In addition, the shipping vial, v-vial,
one day prior to the calibration date, if the total and syringe should all be appropriately shielded
prescribed activity is substantially greater than 3 during the process to minimize radiation expo-
GBq based on a treatment-planning technique that sure (especially hand β absorbed dose) to the per-
differs from those recommended by the manufac- son performing the dispensing. (It may be possible
turer.) An administration kit, infusion v-vial, and to wait to transfer the activity from the syringe to
acrylic shield for each dosage to be dispensed are the v-vial until it has been determined that the
shipped along with the activity but the licensee is correct amount of activity has been withdrawn
responsible for dispensing each of the one or more from the shipping vial into the syringe. However,
7.3 Dosage delivery to and preparation in the hot lab / 7.3.2 Thereasphere dosage dispensing 145

(a)

(b)

Figure 7.3 SIR-Spheres manufacturer’s dose vials. (a) A ten milliliters glass shipping vial containing 3
GBq ± 10% at 18:00 U.S. ET and associated lead pot and (b) patient dose delivery v-vial containing
dispensed target volume activity and associated acrylic shield.

caution must be observed with such an approach 7.3.2 THERASPHERE DOSAGE


to ensure the procedure is performed quickly DISPENSING AND
enough to avoid the aggregation of microspheres PREPARATION
at the bottom of the syringe, which could impede
the transfer of the entire contents of the syringe The activities of all TheraSphere dosages are cali-
to the v-vial.) A detailed step-by-step procedure brated for Sunday at 12:00 U.S. ET and each dosage
for dispensing SIR-Spheres dosages is provided in has a treatment shelf-life of 12 days postcalibra-
the manufacturer’s package insert (Sirtex Medical tion (BTG International Ltd., 2010, 2014; Dezarn
Limited, 2010). An example dispensing worksheet et al., 2011). Originally, the manufacturer only
is shown in Figure 7.4. allowed the ordering of dosages with calibrated
146 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

Figure 7.4 Example SIR-Spheres patient dose dispensing worksheet. The amount of activity required
for each SIR-Spheres patient dose must be transferred from the manufacturer’s shipping vial to the
delivery v-vial based upon the computed activity concentration at the time of dispensing.

activities of 3, 5, 7, 10, 15, or 20 GBq. Dosages later increased that up to 12 days to allow what
may now be ordered with activities between 3 and it calls extended shelf-life treatment, whereby an
20 GBq in 0.5 GBq increments. Also originally, enhancement of the embolic effect for the same
each dosage had a treatment shelf-life of only 5 activity delivered to the same treatment volume
days postcalibration. However, the manufacturer may be achieved. The enhancement is achieved by
7.3 Dosage delivery to and preparation in the hot lab / 7.3.2 Thereasphere dosage dispensing 147

ordering the dosage at a much higher calibrated computed and ordered. The manufacturer provides
activity 1 week earlier and having it decay longer a so-called “treatment window illustrator” elec-
until it reaches the treatment activity level result- tronic spreadsheet, where treatment target volume,
ing in an infusion of a much larger number of desired absorbed dose, variance from U.S. ET, %
spheres for the same infused activity. lung shunt, and anticipated % waste are entered;
Preparation of TheraSphere dosages is and tables of estimated treatment target absorbed
much simpler than that for SIR-Spheres (BTG doses are computed for specific times on each day
International Ltd., 2010; Dezarn et al., 2011). of the first or second week postcalibration for each
During the planning phase, the treatment is sched- allowed noon Sunday U.S. ET calibrated activity
uled for a particular day of the week and the activ- (Figure 7.5). The calibrated activity for which those
ity required on the date of calibration (Sunday of calculated absorbed doses are within the range of
that week or the week before) that will decay to the prescribed absorbed dose on the planned day of
the prescribed activity on the day of treatment is treatment (i.e., within the treatment time window)

Figure 7.5 Top portion of the TheraSphere manufacturer-provided treatment window illustrator
spreadsheet used for determining how much activity calibrated at Sunday noon U.S. ET to order given
a specified target absorbed dose and scheduled day of treatment postcalibration.
148 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

Figure 7.6 TheraSphere patient dose delivery Figure 7.7 Setup for pretreatment ion cham-
v-vial in its acrylic shield and associated lead pot. ber survey meter exposure rate reading of a
TheraSphere dose delivery v-vial, using the fixed-
is the one that is ordered. Each ordered dosage is geometry template provided by the manufac-
then delivered prior to or on the day of treatment, turer (30 cm v-vial center-to-ion chamber center
with the microspheres already in 0.6 mL of ster- distance).
ile water in the infusion v-vial, sealed in its acrylic
A linear regression analysis of exposure rate versus
shield, and ready for administration to the patient
dosage vial activity for the first 20 or so measure-
(Figure 7.6). Thus, the only dosage “preparation”
ments may be useful (Figure 7.8) to derive a dosage
required is activity assay (and exposure rate mea-
vial exposure rate constant (e.g., mR/mCi-h, μSv/
surement, which is described in the next section). GBq-h) that can then be used for comparing future
dosage vial measurements against an expected
7.3.3 EXPOSURE RATE value (exposure rate constant times assayed activ-
MEASUREMENT OF DOSAGE ity) to assess whether or not the measurement was
VIALS made correctly (Erwin, 2012; Gress and Erwin,
2015). A geometric template for survey meter mea-
A measurement of the exposure rate or dose equiv- surement of both the dosage vial and radioactive
alent rate (e.g., mR/h, μSv/h) from each dosage vial waste container has been developed for one of the
in its acrylic shield using a calibrated ion chamber radioembolization products (Figure 7.7).
survey meter and a fixed geometry is required for
both radioembolization products (Figure 7.7). This
measurement is necessary in order to estimate
7.3.4 DOSAGE CART PREPARATION
the infusion waste activity fraction and subse- After each treatment dosage has been prepared,
quently net activity administered as the only way assayed, and had its exposure rate measured,
to “assay” the waste is by survey meter measure- the components of the device apparatus for each
ment (see Section 7.7.1), the measurement of which treatment dosage are placed on a separate cart
is then compared with that of the dosage vial to for transport from the hot lab to the IR proce-
compute the estimated fraction. Furthermore, this dure room. The top surface should be sterilized
measurement must be made correctly as it is the and draped with fresh chux to absorb any spilled
only basis upon which a possible misadministra- or leaked microspheres. The SIR-Spheres infu-
tion (net administered activity or absorbed dose sion system consists of the delivery box contain-
outside of ±20% of prescribed) can be detected. An ing the dosage v-vial in its acrylic shield placed
incorrect survey meter measurement of the dosage in the retaining ring and both the acrylic shield
vial results in a corresponding incorrect estimated cap and delivery box lid on and the dosage deliv-
net administered activity (and by way of direct pro- ery set (infusion lines plus needles) in its sterile
portionality, net absorbed dose) and could result in package (Figure  7.9a). The TheraSphere infusion
either a missed or false-positive misadministration. system consists of the delivery box containing the
7.3 Dosage delivery to and preparation in the hot lab / 7.3.4 Dosage cart preparation 149

External exposure rate vs. SIR-Spheres vial activity


5.00
4.50

Exposure rate @ 30 cm (mR/h)


4.00
y = 0.0584x
3.50 r = 0.995
(n = 40)
3.00
2.50
2.00
1.50
1.00
0.50
0.00
0 10 20 30 40 50 60 70 80 90
Radioactivity in SIR-Spheres delivery vial (mCi)
(a)

External exposure rate vs. TheraSphere vial activity


14.00

12.00
Exposure rate @ 30 cm (mR/h)

10.00 y = 1.682x
r = 0.993
(n = 57)
8.00

6.00

4.00

2.00

0.00
0.00 1.00 2.00 3.00 4.00 5.00 6.00 7.00 8.00
Radioactivity in TheraSphere delivery vial (GBq)
(b)

Figure 7.8 Graphs of fixed-geometry ion chamber survey meter exposure rate reading of 90Y micro-
spheres dose delivery v-vial in acrylic shield as a function v-vial activity for (a) SIR-Spheres and (b)
TheraSphere. An exposure rate constant (e.g., mR/mCi-h) can be computed as either the slope of a
linear regression of measured exposure rate versus activity or the statistical average ratio of expo-
sure rate to activity, and then used to compare the actual reading of each subsequent vial with an
expected reading to ensure a proper reading for the later estimation of fraction of total v-vial activity
delivered to the patient.

dosage v-vial in its lead pot placed inside in the (SIR-Spheres) or lead pot (TheraSphere) and the
retaining ring and both the lids (delivery box, lead delivery box, containing
pot) on a solid-state electronic dosimeter attached
to its holder on the outside of the delivery box 1. Patient name and medical record number
and the administration kit (infusion lines, needle 2. Procedure (e.g., radioembolization)
plunger assembly, and pressure-relief valve and 3. Radionuclide (90Y)
vial) in its sterile package (Figure 7.9b). Duplicate 4. Product (SIR-Spheres or TheraSphere)
labels should be affixed to both the acrylic shield 5. Activity (mCi or GBq)
150 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

(a)

(b)

Figure 7.9 Yttrium-90 microspheres dose administration kits. (a) SIR-Spheres (infusion lines and
needles) and (b) TheraSpheres (infusion lines, needle plunger assembly, infusion syringe, and
pressure-relief valve and vial).

6. Anatomical treatment target (e.g., R Lobe) lower shelves of the cart are a (preferably pancake)
7. Dates and times of assay (and by whom), probe Geiger–Mueller (G–M) survey meter; a cali-
anticipated infusion, and expiration brated ion chamber survey meter (if monitoring of
exposure or dose equivalent rate from the patient
This will help to ensure the correct dosage is will be performed); a β detector survey meter (for
administered to the correct anatomical target in TheraSphere); a 2-L Nalgene waste container (jar),
the correct patient. Fresh, sterile gloves should and an associated acrylic β shield for storing the
be worn while assembling and transporting the residual activity in the infusion system (one per
cart; and once assembled, the cart may need to treatment dosage); and one or more biological
be covered with plastic if being transported to a waste trash bags (Figures 7.10 and 7.11). (Labeling
sterile IR suite. Additional items to place on the each waste jar prior to transport with “90Y,”
7.4 Radiation safety during treatment in the IR suite / 7.4.1 Immediate preadministration 151

Figure 7.10 Radiation detectors employed during 90Y microspheres radioembolization. From left to
right: pancake probe G–M survey meter (protective cover removed for detecting β particles); β probe
survey meter with plastic attenuator (for measuring count rate from TheraSphere microspheres at the
output line–microcatheter junction); electronic personal dosimeter (for measuring the external brems-
strahlung dose rate from the TheraSphere vial); and ion chamber survey meter (for measuring external
bremsstrahlung exposure rate from the patient’s liver containing microspheres).

“SIR-Spheres,” or “TheraSphere,” date, and ana-


tomical treatment target is advisable, so that only
estimated activity and time of day need to be added
after postprocedure assay.) Each treatment dosage
can be kept behind lead shielding and placed in the
delivery box only after IR informs the hot lab that
the IR physician is almost ready to infuse in order
to reduce the exposure to hot lab personnel.

7.4 raDIatION SaFEtY DUrING


trEatMENt IN tHE Ir SUItE

7.4.1 IMMEDIATE
PREADMINISTRATION
The treatment dosage should be requested and the
dosage cart assembled and transported to the IR
suite, shortly before the IR physician has completed
the embolization of any nontarget vessels into which
microspheres could potentially be diverted and
positioned the microcatheter in the desired target
treatment location in the vasculature. A just-in-
Figure 7.11 TheraSphere treatment delivery cart. time delivery of treatment dosages, as opposed to
Top shelf: dose v-vial in acrylic shield and lead transporting them to the IR suite well in advance
pot, inside the infusion delivery box, and dose
of the radioembolization procedure, has the advan-
administration kit. Middle shelf: biohazard bag
for collecting potentially radioactive garments tages of both reducing exposure to IR personnel
and surface coverings, and a 2-L Nalgene waste who are unaccustomed to being in the presence of
jar inside its shield. Bottom shelf: β scintillator, sources of radioactivity and minimizing the possi-
pancake G–M, and ion chamber survey meters. bility of radioactive sources being left unattended in
152 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

a potentially unsecured area. If the IR suite is within covers, gowns, and caps must be worn by any
a defined sterile zone, a sterile plastic cover should personnel who could come in direct contact with
be placed over the dosage cart once it is assembled microspheres (most likely the physician admin-
for transport and kept in place until the cart is ready istering the dosage, but also potentially anyone
to be wheeled into the procedure room. directly assisting with the infusion). The fluoros-
The first step in a radioembolization is a time- copy system should be covered with disposable
out prior to the dosage cart being wheeled into the clear plastic and disposable absorbent material
procedure room to ensure the correct dosage will be should be placed over the patient and attached to
administered to the correct treatment target in the the floor, with a minimum of 6 ft × 6 ft area of floor
correct patient. Once that has been verified, the cart coverage recommended (Salem and Thurston,
is wheeled into the procedure room and into posi- 2006), where spilled or leaked microspheres have a
tion for infusion (as close as possible to the fluoros- high likelihood of landing.
copy table, with the delivery box oriented such that
its output side is on the patient side of the cart) by 7.4.2 DURING ADMINISTRATION
the NM technologist or physicist. Towels or absor-
bent pads should then be placed between the dosage Once assembly of the infusion system has been
cart and patient entry point with overlap to confine completed and verified, the physician begins to
any spill or leakage of microspheres along the path administer the microspheres to the patient. The
between the output side of the delivery box and fem- system should be visually monitored at the start
oral artery entry point of the microcatheter. Next, of infusion, and then periodically throughout, to
the input and output lines plus needles are removed ensure that the system remains “closed” (i.e., there
from their package and primed with sterile water is no leakage of fluid, especially around the septum
or saline according to the manufacturer’s instruc- of the v-vial and at the output line–microcatheter
tions to ensure that there is no air in the infusion junction). Leakage from the septum of the SIR-
lines. Finally, the infusion system is assembled, Spheres v-vial can be visualized directly as the
again according to the manufacturer’s instruc- v-vial is only shielded with transparent acrylic,
tions. Developing a checklist with time-outs at whereas the TheraSphere v-vial in its acrylic shield
various steps in the process is highly recommended is kept within a lead pot and the needle plunger
(Figure 7.12) to help ensure that all assembly steps is attached to the acrylic shielding so that what is
are correctly followed (Salem and Thurston, 2006). monitored instead is leakage from the seal around
Radioembolization is typically performed infre- the plunger connection to the acrylic shield. An
quently and is a unique form of brachytherapy ion chamber survey meter may be held externally
that differs substantially from routine NM and IR above the patient’s liver (at approximately 6 in.) for
procedures. Furthermore, there will be turnover of real-time verification that microspheres are being
NM and IR personnel participating in this proce- delivered to the liver. (The exposure rate or dose
dure over time. These factors increase the likelihood equivalent rate reading should increase as the infu-
of mistakes that could be detrimental to either the sion progresses. Note, however, that this form of
patient or personnel if all the required steps in the verification is limited, as such meters tend to be
process are not adhered to strictly (i.e., misadminis- omnidirectional and are thus unable to localize
trations or contaminations) and justify the use of a the direction of the source of radiation with high
checklist even if certain personnel believe that they accuracy and precision.)
have mastered the procedure. SIR-Spheres dosage administration consists of
Since radioembolization involves RAM and is sequences of syringe-pulsed infusions of micro-
performed under fluoroscopic guidance, all per- spheres (0.25–0.5 mL per pulse, at a rate of not
sonnel in the room during the procedure must more than 5 mL per minute) using sterile water,
wear appropriate radiation protection garments with periodic pauses for contrast fluoroscopy to
(lead aprons and collars, protective eyewear) and ensure that flow to the target is being maintained
whole-body dosimeters (and ring dosimeters and the location of the microcatheter has not
for anyone who might receive measurable hand changed (Sirtex Medical Limited, 2003; Salem and
exposure from fluoroscopy x-ray or microsphere Thurston, 2006). A control knob on the delivery
β-radiation). In addition, disposable surgical shoe box allows the physician to toggle the input to the
Figure 7.12 Example TheraSphere dose administration checklist. Checklists for (1) items required; (2) documentation required; (3) preadministration
activity measurements; (4) cart preparation; (5) postadministration; (6) residual measurement; and (7) radiology information system (RIS) records and
documentation are also contained in the document from which the checklist shown was obtained.
7.4 Radiation safety during treatment in the IR suite / 7.4.2 During administration 153
154 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

three-way stopcock on the output side between the of the acrylic delivery box measures the external
output line of the v-vial and the contrast/sterile dose equivalent rate from the lead pot containing
water line. Stasis is a common occurrence with the dosage vial. An initial reading just before the
SIR-Spheres due to complete embolization of the start of infusion is recorded, and a second recorded
target as a consequence of the large number of when the physician has stopped the infusion. The
microspheres involved. A temporary loss of ante- second reading should be zero or a very small frac-
grade flow may also occur due to vascular spasms tion (<0.05) of the initial reading under normal
in reaction to the infusion of the microspheres. circumstances. (The readings must be recorded
Infusion must not continue if stasis has occurred only when the fluoroscopy x-ray beam is off and
or until antegrade flow has been restored as inad- the dosimeter reading has stabilized. The scattered
vertent delivery of microspheres to nontarget tis- x-ray radiation will cause the dosimeter reading to
sues via arteries upstream from the position of the increase substantially and a 5- to 10-second delay
microcatheter may occur, causing a misadminis- after the x-ray beam is turned off before recording
tration (and possibly a reportable medical event), the reading is required due to the slow response
as well as possibly causing radiation-related nor- time of the dosimeter.) The second method of veri-
mal tissue complications (e.g., ulcerations) requir- fication is the counts per minute (cpm) reading of
ing medical intervention. The infusion proceeds a β detector (thin-wafer plastic scintillator insensi-
until either the entire dosage has been adminis- tive to gamma and x-ray radiation) survey meter
tered, stasis has been reached, or early termination in close proximity to, and directed at, the infusion
occurs due to an emergent patient condition (e.g., system output line/microcatheter junction after the
an unacceptable level of abdominal pain). An entire initial 20 mL of flush. The initial high cpm reading
SIR-Spheres dosage has essentially been infused should stabilize at a much lower reading after two
when the fluid in the v-vial has transitioned from or more follow-on 20 mL flushes. (Depending on
its initial sandy-yellow translucent color to being the sensitivity of the β detector, a plastic attenua-
almost clear. An air-filled syringe can be used to tor over the entrance window may be required to
flush the remaining fluid from the v-vial. A single achieve counts per minute (cpm) readings within
SIR-Spheres dosage administration may take up to the lower range settings of the survey meter.) The
20 minutes due to the slow nature of the infusion reading will not decrease to zero as some of the
process. microspheres, albeit a small fraction, will inevi-
Administration of a TheraSphere dosage occurs tably become attached to or trapped in the v-vial,
much more rapidly than that for SIR-Spheres output line, microcatheter, and, in particular, the
(Salem and Thurston, 2006; BTG International output line–microcatheter junction. Periodic gen-
Ltd., 2010, 2014). Twenty milliliters of fluid is tle tapping on the output line–microcatheter fitting
drawn from a ≥100-mL saline bag attached to the with a hemostat during flushing can free up some
infusion system and into the integrated 20-mL of the microspheres that have become trapped at
syringe and then flushed through the v-vial using that point.
steady pressure (≤30 psi). (A relief valve that
diverts fluid into a vented 20-mL vial is integrated
into the input line to the v-vial, should the pressure 7.4.3 IMMEDIATE
exceed 30 psi.) One-way valves in the lines between POSTADMINISTRATION
the saline bag and syringe and syringe and v-vial
are incorporated to prevent reverse flow of fluid. After the administration of each treatment dos-
The TheraSphere system is designed to infuse the age has either been completed or terminated early
vast majority of the microspheres into the patient due to stasis or emergent patient condition, the
with the initial 20 mL of flush and infuse nearly radioactive items from the infusion must be segre-
all of the remaining microspheres via a minimum gated and placed in the Nalgene radioactive waste
of two additional 20 mL of flushes. Two methods container for later measurement of the estimated
are employed for verification that the TheraSphere fraction of the total activity that was not delivered
infusion has been completed (Salem and Thurston, to the patient for that particular treatment (see
2006; BTG International Ltd., 2010, 2014). First, Section 7.7.1). Since the input and output lines,
the electronic dosimeter attached to the input side needles (and needle plunger for TheraSphere),
7.5 Patient release / 7.5.1 To the public 155

dosage vial in its acrylic shield, and microcath- Personnel who were in the procedure room dur-
eter have all been interconnected, they are to be ing the radioembolization(s) and who could pos-
treated as a single radioactive item at the end of sibly have been contaminated with microspheres
infusion (as opposed to being disassembled). must be surveyed head-to-toe with the G–M survey
The SIR-Spheres input lines should be recapped meter before leaving the room (Dezarn et al., 2011).
after removing the attached syringes, and the If any radioactivity is detected on protective cover-
TheraSphere input line is cut immediately distal ings (cap, gown, gloves, shoe covers), they must be
to the pressure-relief valve. The dosage vial in its removed and placed in the designated decay-in-
shield followed by the infusion lines should then storage biohazard bag. Afterward, a microspheres
be placed in the waste container first. The infu- contamination survey of the room should be per-
sion microcatheter is then slowly withdrawn from formed, paying particular attention to the coverings
the patient. Coiling up the microcatheter on the over the patient, fluoroscopy system, and floor used
towel or absorbent pad underneath the base cath- during the procedure, and anything containing fluid
eter as it is being removed is recommended, and or blood (Dezarn et al., 2011). (This survey may have
as soon as the tip of the microcatheter appears, it to be performed when the patient is no longer in the
is prudent to clamp it with a hemostat (although procedure room, if the bremsstrahlung radiation
covering it with gauze has also been suggested). emanating from the patient would mask detection
(The same hemostat could be used for cleaning of low levels of β radiation from microsphere con-
the dosage vial septum, tapping the TheraSphere tamination.) Alternatively, the coverings used could
output line–microcatheter junction, and clamp- be carefully folded inward together to confine any
ing the microcatheter.) Blood leaking from the microspheres and placed in a receptacle that could
base catheter should be allowed to drip onto the be later surveyed and sequestered for decay-in-
same towel or absorbent pad before wrapping it storage if found radioactive. If any surfaces in the
around the microcatheter and hemostat and plac- room are found to be radioactive, the institution’s
ing all of them in the waste container. (If a syringe Radiation Safety Officer (RSO) should be contacted
is used to withdraw blood from the base catheter for guidance and oversight regarding attempts at
after removal of the microcatheter, it should be removal and/or cover up of the source(s) of radiation.
considered radioactive and placed in the waste If radiation exposure to personnel is deemed exces-
container.) Other items to be assumed radioactive sive and removal is not possible, then acrylic (≥0.5
are the removed acrylic shield plug, the hemostat inch) should be placed over the source(s) of radia-
and alcohol swab(s) used to clean the dosage vial tion to absorb essentially all of the β particles from
septum, and any gloves that may have come into 90Y until decay to background or a level considered

contact with microspheres (all to be placed in the safe (Dezarn et al., 2011). (Secondary bremsstrah-
waste container), as well as the acrylic delivery lung radiation will be generated in the acrylic but
box. Items on the input side of the infusion sys- it should constitute a negligible fraction of the total
tem that would not come into contact with micro- energy of the incoming β radiation.)
spheres under normal circumstances, and can
thus be considered nonradioactive, are used ster-
ile water, saline, and contrast syringes; towels and/
or absorbent chux; packaging materials; and the 7.5 PatIENt rELEaSE
TheraSphere saline bag, pressure-relief vial, and
infusion line up to where it was cut (as well as the Radioembolized patients will be a source of radia-
scissors used to cut the line). The chux draping the tion exposure above background for an extended
surface of the dosage cart should be surveyed with period of time postinfusion. The duration will be
the G–M meter, and if found to be radioactive, related to both the total infused activity and half-
placed in the waste container. (The entrance win- life of 90Y as the internalized activity decreases
dow of the G–M detector must be exposed for sur- by physical decay only (i.e., there is no biological
veying surfaces for 90Y microspheres. Otherwise, clearance). Thus, the external radiation exposure
most of the β particles will be absorbed by the pro- of others is of concern, in particular, those individ-
tective cover, resulting in either underestimating uals for whom the exposure would be unexpected,
or missing contaminations.) whether they are members of the general public
156 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

(when the patient is released from the licensee’s 0.346 × Γ × Q0 × Tp × E


TEDE ( mSv ) = (7.11)
control) or hospital personnel categorized as non- r2
radiation workers (if the patient needs to be admit-
ted following treatment). where 0.346 = 0.01 mSv/mrem × 24 hours per
day/ln(2); Γ is the radionuclide exposure rate con-
stant (mR-m2/mCi-h); Q0 is the amount of activ-
7.5.1 TO THE PUBLIC ity remaining in the patient at a proposed time of
release (mCi); Tp is the radionuclide half-life (d); E is
The NRC allows the release of a radioactive patient an occupancy factor (0.25 for Tp greater than 1 day);
from radiation confinement as long as the estimated r is the distance (m) from the patient; and a Γ-to-
cumulative (total) radiation exposure to any other EDE rate (mrem/mR) conversion factor of unity is
individual does not exceed 5 mSv effective dose assumed. As stated earlier, 90Y is a pure beta emit-
equivalent (EDE). However, if the estimated maxi- ter. Thus, the only external radiation from the
mum EDE exceeds 1 mSv, then verbal and written patient would be that due to secondary bremsstrah-
instructions must be provided to the patient and a lung x-rays produced that are not absorbed by the
record of them maintained by the licensee in order patient. A published value for the bremsstrahlung
to keep the exposure as low as reasonably achiev- exposure rate constant for 90Y based on uniform
able (ALARA) (NRC, 2013; Siegel, 2004). The only distribution in a reference adult model is 5.64 × 10–4
exposure others will normally receive from a patient mR-m2/mCi-h (Zanzonico et al., 1999) and values
to whom permanent radioactive implants have been of 1.84 ± 0.53 × 10–3 and 2.82 × 10–3 mR-m2/mCi-h
administered is external (as opposed to a radiophar- have been obtained from measurements at 1 m from
maceutical such as iodine-131 sodium iodide, which radioembolization patients (Figure 7.13) (Gulec and
must take into account potential internal exposure Siegel, 2007; Erwin et al., 2012). Assuming the worst
due to intake of activity that is excreted). case, the highest of the three Γ values for 90Y, the
Radioembolization is considered a type of per- amount of infused activity at which release instruc-
manent implant. The equation governing the esti- tions would be required (TEDE = 1 mSv), would be
mation of the total EDE (TEDE) received by the
most exposed person for the case of permanent 0.037 GBq / mCi × 1 mSv
57 GBq = (7.12)
implants is (NRC, 2008) 0.346 × 2.83E − 3 × 2.67 d × 0.25

Exposure rate vs. 90Y microspheres activity


0.500
0.450
y = 0.00176x
Exposure rate @ 1 m (mR/h)

0.400
r = 0.876
0.350 (n = 20)
0.300
0.250
0.200
0.150
0.100
0.050
0.000
0.0 50.0 100.0 150.0 200.0 250.0
90Y microspheres activity (mCi)

Figure 7.13 Graph of measured posttreatment external bremsstrahlung exposure rate versus net
administered activity for 20 90Y microspheres patients (9 SIR-Spheres, 11 TheraSpheres). The slope of
the linear regression or the statistical average ratio of exposure rate to activity represents a measured
90Y microspheres patient bremsstrahlung exposure rate constant.
7.6 Significant poasttreatment events / 7.6.2 Autospy, burial, or creamation 157

and the activity above which a period of radia- no requirement to place them in radiation con-
tion confinement before release is required would finement under the care of personnel trained in
be five times that amount. The theoretical upper radiation safety and allowed to receive exposures
limit on the infused activity for a single radio- above that to members of the public (including
embolization treatment is currently 20 GBq and nonradiation hospital workers). However, one
the typical infused activity is roughly an order may choose to do so if the patient is admitted
of magnitude below that resulting in 1 mSv soon after treatment and will require intensive
TEDE. Thus, all radioembolization patients may care, whereby personnel will spend a substan-
currently be released immediately without the tial amount of time at close proximity to the
documentation of verbal and written instruc- patient and it is deemed possible that they will
tions. However, it is prudent to provide all such receive substantially higher than general public
patients with documentation indicating that limit exposures. A standardized document could
they are radioactive and for how long, espe- be developed and provided with each patient to
cially if they intend to travel soon enough after notify the hospital staff caring for the patient
the treatment to potentially trigger radiation that universal precautions should be observed
detectors at an airport or border crossing. Trace while caring for a 90Y microspheres patient while
amounts of unbound 90Y from SIR-Spheres have he or she is still radioactive (Figure 7.14). Such a
been detected in urine (25–50 kBq/liter per GBq) document is especially useful when patients are
within the first 24 hours after implant (Sirtex admitted to a regular room where personnel are
Medical Limited, 2003; Lambert et al., 2011). not trained as radiation workers. If a SIR-Spheres
Although not considered a significant source of patient requires urinary catheterization requir-
radiation exposure, all patients could be asked ing the changing of collection bags, then it would
to flush twice after urination and male patients be prudent for the personnel handling them to be
asked to sit during urination for 1 day after treat- gloved and empty the bags into the patient’s toilet
ment (Dezarn et al., 2011). followed by two flushes until the radiation level
in the bag reaches background. If any radioembo-
lized patient requires abdominal drainage, then
7.5.2 HOSPITAL ADMISSION an assessment of the radiation level in the drain-
age bag should be made, and if radioactive then
As the above 90Y radioembolization patient medical intervention may be indicated, as under
release calculation demonstrates, if a patient must normal circumstances there should be no radio-
be admitted after treatment, there is currently activity in the abdominal fluid.

Figure 7.14 Example notification of 90Y microspheres patient, should he or she need to be admitted
to the hospital while still considered radioactive. Such patients can be released to the general public
immediately without radiation precaution instructions. However, if they need inpatient care, prudence
suggests that hospital staff caring for the patient be made aware that he or she is radioactive and
what precautions, if any, are necessary.
158 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

7.6.2 AUTOPSY, BURIAL, OR


7.6 SIGNIFICaNt
CREMATION
POSttrEatMENt EVENtS
Individuals currently treated with TheraSphere or
7.6.1 SURGERY: LIVER RESECTION SIR-Spheres tend to be late-stage cancer patients.
OR TRANSPLANT, OR OTHER Thus, there exists a possibility of a patient expiring
posttreatment while still radioactive. As explained
One of the clinical indications of TheraSphere above (see Section 7.5), radioembolization patients
is radiotherapy neoadjuvant to liver resection may be released to the general public without radi-
or transplantation. Therefore, a percentage of ation precaution instructions as the most exposed
patients treated with TheraSphere will proceed person is estimated to receive an exposure about
to surgery. If surgery is planned for a radioem- an order of magnitude lower than the limit for a
bolized patient, then how long before resection member of general public. The exposure to others,
or transplant can occur from a radiation safety including embalmers and funeral workers, during
standpoint must be a consideration. A surface body preparation, visitation, funeral, and/or burial
dose equivalent rate of 20 μSv/h above the liver is following the death of a radioembolization patient
a generally accepted threshold below which radia- is anticipated to be even lower than that to the
tion safety precautions are not required for sur- most exposed person under normal circumstances
gery postradioembolization (Salem and Thurston, (all these individuals being exposed over a much
2006). A posttreatment surface dose equivalent shorter time period overall), and thus should not be
rate measurement combined with the physical of concern (Dezarn et al., 2011). Furthermore, no
half-life of 90Y can be used to predict when sur- special precautions are required during embalming
gery can be performed without concern regarding using standard methods. Microspheres are perma-
the radiation exposure to personnel (in particu- nent implants, and thus there will be no radioac-
lar, the surgeon). The explanted liver tissue should tivity in the aspirated blood (aside from possible
be placed in a leakproof container contain- trace amounts of 90Y dissociated from SIR-Spheres,
ing formaldehyde and refrigerated and located which has been detected in urine and which sug-
behind lead shielding for radioactive decay-in- gests it was present in blood and extracted by the
storage if the dose equivalent rate at the surface kidneys).
of the container is greater than 50 μSv/h (Salem If an autopsy is to be performed, then depend-
and Thurston, 2006). Surgery due to an emergent ing upon how long after infusion of the micro-
condition while the liver is still considered radio- spheres the autopsy is scheduled, it may be
active should not be prohibited as the welfare prudent for the pathologist to explant the liver
of the patient outweighs an infrequent and not and have it relocated for radioactive decay-in-
excessive personnel radiation exposure. Under storage before proceeding with the autopsy on
all circumstances, the institution’s RSO should the remainder of the body to minimize unin-
be consulted to provide guidance related to the tended exposure. According to the recommen-
surgery itself (e.g., the use of lead surgical gloves dations found in International Commission on
and other radioprotective apparel and body and Radiological Protection publication 94 (Harding
extremity dosimeters); handling of the explanted et al., 2004), routine autopsy procedures may be
liver tissue (by both surgery and pathology) and followed if a corpse contains less than 0.45 GBq
any other items that may be radioactive as a result of sealed 90Y; but if still considered a radiation
(e.g., blood, surgical garments and instruments, hazard and the radioactivity is confined to a spe-
and towels and other surface-covering materials); cific organ, then that organ should be explanted
surveys of all personnel and areas where RAM and stored for radioactive decay.
contaminations may have occurred; labeling of Cremation of a corpse containing radioac-
all radioactive items; and appropriate signage in tive microspheres presents a special case radiation
the surgical suite and other areas where RAM hazard, with the possibility of not only external
is present (Salem and Thurston, 2006; Dezarn exposure but also internal exposure due to the inha-
et al., 2011). lation of RAM in the residual ashes or crematorium
7.7 Radioactive waste / 7.7.2 Example procedure measurements and calculations 159

effluent (NCRP, 2006; Nelson et al., 2008; Dezarn microcatheter, the towels or absorbent pads under
et al., 2011). The radiation hazard is complicated by the base and microcatheters (and any syringe into
the presence of long-lived impurities in the micro- which radioactive blood was withdrawn from the
spheres (see Section 7.7, “Radioactive Waste”). The base catheter), the acrylic shield plug, the alco-
limit on the amount of 90Y radioactivity in a body hol swab used to clean the dosage vial septum,
to be cremated varies country by country, with val- hemostat(s) used to wipe the septum and clamp
ues ranging from 0.047 GBq in the United States the tip of the microcatheter postinfusion, and the
to 1 GBq in Australia (Harding et al., 2004). Local acrylic delivery box (and elsewhere if a contami-
regulations regarding limits on radioactivity in the nation or spill occurs) (Salem and Thurston, 2006;
effluent from the cremation process may also apply Dezarn et al., 2011). Therefore, a direct measure-
(Nelson et al., 2008). The ideal scenario is to have the ment of the residual activity with a dose calibra-
liver explanted (and possibly the lungs as well, in the tor is not possible. Instead, an estimate must be
case of substantial shunting of microspheres) and derived from calibrated ion chamber exposure
stored for the radioactive decay prior to presenting rate or dose equivalent rate readings of the brems-
the body for cremation (Dezarn et al., 2011). If the strahlung radiation from the dosage vial prior to
liver is not explanted (nor are radioactive lungs), administration and the waste container (inside its
then the body will have to be stored for a period of acrylic shield) afterward, using a fixed measure-
time before cremation, if the on-board activity at the ment geometry (Figure 7.15) (Salem and Thurston,
time of death exceeds the local regulatory limit. 2006; BTG International Ltd., 2010). The dosage
General recommendations regarding handling vial is essentially a radially symmetric radiation
of radioactive patients post-mortem can be found source, so only a single reading from any direc-
in National Council on Radiation Protection tion is necessary. However, the residual activity
Report Nos. 155 and 161 (NCRP, 2006, 2010), in within the waste jar is not isotropic; therefore, an
addition to ICRP publication 94. The institution’s average of readings for multiple rotations of the
RSO should be consulted for guidance specific acrylic shield in front of the survey meter (typi-
to radioembolized patients, regarding radiation cally four, at 0°, 90°, 180°, and 270° of rotation)
safety precautions related to the corpse, excised
liver (and possibly lungs), and potentially radio-
active blood or urine, and to ensure compliance
with local regulations. Finally, depending upon
whether the death occurs inside or outside of the
institution where the patient underwent the pro-
cedure, either a physician or an RSO involved or a
family member of the deceased should inform the
morgue, funeral home, and/or crematorium that
the decedent underwent a radioembolization pro-
cedure and when. Those entities can then consult
with the treating institution’s AU or RSO to assess
whether or not a radiation hazard is presented at
the various stages post-mortem (NCRP, 2006).

Figure 7.15 Setup for TheraSphere posttreatment


7.7 raDIOaCtIVE WaStE ion chamber survey meter exposure rate read-
ing of residual activity, using the fixed-geometry
template provided by the manufacturer (30-cm
7.7.1 RESIDUAL ACTIVITY ASSAY waste jar center-to-ion chamber center distance).
The average of four readings, with the waste jar
As mentioned in Section 7.4.3, the residual activ- plus shield rotated 90° between each, is com-
ity for each radioembolization treatment dosage pared with that from the v-vial in shield prior to
will reside not only in the dosage vial but can treatment, to estimate the fraction of total v-vial
also reside in the infusion needles and tubing, the activity delivered to the patient.
160 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

is computed. (A background reading is also Average residual waste R: 0.0852 mR/h (decayed
obtained at the time of, and subtracted from, the to time of infusion)
dosage vial and waste container readings.) The Waste ratio W: 0.013
two measured rates (R) are decay corrected to the Net activity administered:
time of infusion and the estimated residual activ- 4.16 GBq = 4.21 × (1 − 0.013)
ity is then calculated:
Percent of Dtumor(Gy) delivered:
residual  Aresidual (GBq) =  A(GBq) × W (7.13) 4.16 GBq
93.9 % = 100 × (Dtumor(Gy) ∝ A(GBq))
4.43 GBq
where initial A(GBq) is the preadministration
vial activity calibrator assay of the treatment dos- The estimated absorbed dose delivered to the
age vial (decay corrected to the time of infusion), target was well within the regulatory limit of ±20%
and W is the ratio of measured rates (Rwaste/Rvial). of the prescribed absorbed dose.
A spreadsheet that automates the calculation of Example 7.2: Reportable medical event
residual activity, as well as the estimates of net A patient with hepatocellular carcinoma and
activity administered and absorbed (and percent bi-lobar disease was treated with two dosages of
of prescribed) dose, is illustrated in Figure 7.16. TheraSphere, one for each lobe of the liver. The
right lobe infusion was successful, resulting in
7.7.2 EXAMPLE PROCEDURE the delivery of an estimated 96% of the prescribed
MEASUREMENTS AND 115 Gy absorbed dose. However, the left lobe infu-
CALCULATIONS sion resulted in a reportable medical event due to a
breach of the seal around the plunger assembly vial
The measurements and calculations surrounding interface.
the radioembolization procedure itself that are Target Mliver(kg): 0.755 kg (1.03 kg/L × 0.733 L
used to estimate the net activity administered and estimated using CT)
absorbed dose delivered, as well as assess whether Prescribed Dtumor(Gy): 80 Gy
or not a reportable medical event has occurred, Lung shunt fraction (SF): 0.1035
are illustrated in this section. Two examples are Required A(GBq) for 80 Gy:
provided, both based on actual cases. The first
80 Gy × 0.755 kg
example demonstrates a case where the outcome 1.35 GBq =
of the infusion was as expected. The second exam- (49.98 Gy ⋅ kg / GBq × [1 − 0.1035] )
ple illustrates a misadministration that occurred
as a result of a failure of the device, resulting in a Dispensed A(GBq) assay: 1.32 GBq (decayed to
reportable medical event. time of infusion)
Dosage vial R: 2.19 mR/h (decayed to time of
Example 7.1: Successful infusion infusion)
A patient whose entire right lobe of the liver was Average residual waste R: 0.82 mR/h (decayed
replaced by a pancreatic neuroendocrine cancer to time of infusion)
metastasis was treated with SIR-Spheres based on Waste ratio W: 0.374
a target absorbed dose prescription.
Net activity administered:
Target Mliver(kg): 2.98 kg (1.03 kg/l × 2.893 L
estimated using CT) 0.83 GBq = 1.32 × (1 − 0.374 )
Prescribed Dtumor(Gy): 70 Gy Net absorbed dose delivered:
Lung shunt fraction (SF): 0.0577 49.98 Gy − kg / GBq × 0.83 GBq × (1 − 0.1035)
Required A(GBq) for 70 Gy: 49 Gy =
0.755 kg
70 Gy × 2.98 kg
4.43 GBq = 49 Gy
(49.98 Gy ⋅ kg / GBq × [1 − 0.0577] ) Percent of Dtumor(Gy) delivered: 61 % =
80 Gy
Dispensed A(GBq) assay: 4.21 GBq (decayed to (|Delivered – Prescribed|): 31 Sv (1 Gy = 1 Sv for
time of infusion) electrons and photons)
Dosage vial R: 6.63 mR/h (decayed to time of The estimated absorbed dose delivered to
infusion) the target was less than 80% of the prescribed
7.7 Radioactive waste / 7.7.3 Storage for decay and disposal 161

Figure 7.16 Example TheraSphere day-of-therapy spreadsheet that includes a comparison of mea-
sured and expected exposure rates from the delivery v-vial in acrylic shield, as well as calculations of
net activity and absorbed dose delivered to the patient and comparison to the prescribed dose.
162 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

absorbed dose, and the difference between the esti-


mated absorbed dose delivered and the prescribed
absorbed dose exceeded the threshold for report-
ing a misadministration (0.5 Sv equivalent dose to
an organ or tissue). Thus, reporting this medical
event to the NRC or Agreement State regulatory
authority was required.

7.7.3 STORAGE FOR DECAY AND


DISPOSAL
The radioactive waste from each radioemboliza-
tion procedure requiring a period of decay-in-
storage prior to disposal typically consists of (1)
one or more waste jars containing the residual
infusion system activity (sealed and removed
from the reusable acrylic shield); (2) any biohaz-
ard bags containing microspheres-contaminated
items such as gloves, garments, surface coverings,
and absorbent media employed in the removal of
microspheres from contaminated surfaces; and
(3) any unused treatment doses due to procedure
cancellation or an emergent patient condition.
Each of these sources of radioactive waste must
be appropriately labeled. The label for each should
include

1. Radionuclide (90Y)
Figure 7.17 Example 90Y microspheres residual
2. Product (SIR-Sphere or TheraSphere) waste jar, labeled for decay-in-storage and later
3. Date and time disposal.
4. Activity (mCi or GBq) for each residual activity
waste jar (Figure 7.17) and unused dosage be significantly affected. (Of course, the effect will
depend on the geometry and self-shielding of the
The 90Y in SIR-Spheres is eluted from a 90Sr/90Y container and its contents, and the distribution of
generator and thus there would ideally be no microspheres within, in addition to the amount of
90 Sr impurity.)
radionuclidic impurities in the radioactive waste.
However, trace amounts of 90Sr have been detected Yttrium-90 is activated in TheraSphere
in SIR-Spheres waste due to “breakthrough” dur- microspheres by neutron bombardment. The
ing the elution process, with amounts of 90Sr on microspheres are manufactured with 89Y as a
the order of 3 Bq per GBq of 90Y at the time of constituent of the glass matrix, and 90Y is pro-
manufacturer assay reported (Metyko et al., 2014). duced via the irradiation of the microspheres
As a consequence, SIR-Spheres waste will remain in a nuclear reactor (89Y(n,γ)90Y). TheraSphere
slightly radioactive with both 90Sr and the 90Y it microspheres are known to contain a number
is generating (both decaying with the 28.79 year of gamma-emitting radionuclidic impurities as
half-life of 90Sr) after all of the initial 90Y activity a result of neutron activation of other elements
has essentially decayed away (Figure 7.18). On the within the glass matrix (NRC, 2007; Ostrowski
other hand, the negligible amount of 90Sr impurity et al., 2007; Nelson et al., 2008; Metyko et al.,
suggests that the duration of time over which the 2012). The impurities that have been detected
radiation level external to the source container is have much longer half-lives than that of 90Y. The
distinguishable from background before it may be two most prominent impurities are 88Y (106.6-day
disposed of as regular, biohazard waste may not half-life) and 91Y (58.5-day half-life) due to the
References 163

Theoretical 90Sr/90Y spectrum


0.14
Counts #
# β particles per nuclear transition

0.12
1200
0.10
1000
0.08 800
Total
0.06 Y-90 600
Sr-90
0.04 400

0.02 200
0
0.00 0 400 800 1200 1600 2000 2400
0 500 1000 1500 2000 2500
KeV
Energy (KeV)
(a) (b)

Figure 7.18 Strontium-90 impurity in SIR-Spheres. (a) Theoretical β energy spectrum for 90Sr/90Y.
(b) Liquid scintillation counter β energy spectrum of an aliquot from an unused SIR-Spheres v-vial,
long after the original 90Y radioactivity had decayed to the level of background, demonstrating the
production of 90Y from the decay of on-board 90Sr.

4000000 7.8 CONCLUSIONS


3500000

3000000 Radioembolization is a unique therapeutic appli-


2500000
cation of RAM and as a consequence presents cor-
respondingly unique challenges from a radiation
Counts

2000000
safety perspective. The intent of this chapter is to
1500000 arm the radioembolization team (interventional
1000000 radiologists, medical physicists, nuclear medicine
500000
physicians, and/or radiation oncologists) with a
thorough understanding of the dosage prepara-
0
250 500 750 1000 1250 1500 1750 2000 2250 2500 tion, patient treatment and release, and radioactive
Energy (KeV) waste management radiation concerns specific to
both SIR-Spheres and TheraSphere. All members
Figure 7.19 Radionuclidic impurities in of the interdisciplinary radioembolization team
TheraSphere. A high-purity germanium detector, should be keenly aware of and adhere strictly to
high-resolution γ-ray emission spectrum from
the various regulatory and good radiation safety
an unused TheraSphere v-vial, long after the
original 90Y radioactivity had decayed to the level practice aspects associated with this treatment
of background. A number of γ-emitting impuri- modality. Doing so will help ensure that a licensee
ties were present, most notably 88Y (898 keV and establishes and maintains a radioembolization
1.836 MeV). program that is both safe and effective for patients,
while simultaneously safeguarding personnel and
members of the public against excessive radiation
reactions 89Y(n,2n)88Y and 89Y(2n,γ) 91Y, respec-
exposure.
tively (Figure 7.19). Both produce high-energy
gamma emissions as a result of radioactive decay
and two from 88Y are high yield (898 keV/93.4%, REFERENCES
1.836 MeV/99.4%). Depending upon an institu-
tion’s RAM license and local regulations and Berger, M.J. (1976). MIRD Pamphlet 7:
whether or not the radioactive impurities in Distribution of Absorbed Dose Around Point
the procedure waste are detectable above back- Sources of Electrons and Beta Particles in
ground, the radioactive waste from TheraSphere Water and Other Media. Reston, VA: The
procedures may have to be shipped to an outside Society of Nuclear Medicine and Molecular
entity for decay-in-storage and disposal. Imaging.
164 Radiation safety concerns associated with preparing the dosage, treating and releasing the patient

Braat, A.J.A.T. et al. (2015). 90Y hepatic radio- Ford, K. (1955). Predicted 0+ level of Zr90. Phys
embolization: An update on current prac- Rev 98:1516.
tice and recent developments. J Nucl Med Gress, D., Erwin, W. (2015). SIR-Spheres dose vial
56:1079–1087. activity assay cross-calibration with an outside
BTG International Ltd. (2010). TheraSphere® radiopharmacy. Med Phys 42:3498.
Reference Manual [Accessed February 24, 2011]. Gulec, S.A., Siegel, J.A. (2007). Posttherapy
BTG International Ltd. (2014). Package Insert – radiation safety considerations in radiomicro-
TheraSphere® Yttrium-90 Glass Microspheres. sphere treatment with 90Y-microspheres. J
Available from: http://www.therasphere.com/ Nucl Med 48:2080–2086.
physicians-package-insert/TS_PackageInsert_ Ho, S. et al. (1996). Partition model for estimating
USA_v12.pdf [Accessed August, 2015]. radiation doses from yttrium-90 microspheres
Busse, N., Erwin, W., Pan, T. (2013). Evaluation in treating hepatic tumours. Eur J Nucl Med
of a semiautomated lung mass calculation 23:947–952.
technique for internal dosimetry applications. Harding, L.K. et al. (2004). ICRP publication
Med Phys, 40:122503. 94: Release of patients after therapy with
Carey, J.E. et al. (2012). AAPM report no. 181: unsealed radionuclides. Ann ICRP 34:1–80.
The selection, use, calibration, and quality ICRU. (1984). Report 37: Stopping Power for
assurance of radionuclide calibrators used in Electrons and Positrons. Bethesda, MD:
nuclear medicine. American Association of International Commission on Radiation Units
Physicists in Medicine, One Physics Ellipse, and Measurements.
College Park, MD, 20740-3846. Johnson, O., Johnson, R., Langer, L. (1955).
Cole, A. (1969). Absorption of 20-eV to 50,000 eV Evidence for a 0+ first excited state in Zr90.
electron beams in air and plastic. Radiat Res Phys Rev 98:1517–1518.
38:7–33. Kao, Y.H. et al. (2012). Image-guided personal-
Dezarn, W.A. et al. (2011). Recommendations of the ized predictive dosimetry by artery-specific
American Association of Physicists in Medicine SPECT/CT partition modeling for safe and
on dosimetry, imaging, and quality assurance effective 90Y radioembolization. J Nucl Med
procedures for 90Y microsphere brachytherapy 53:559–566.
in the treatment of hepatic malignancies. Med Kennedy, A. et al. (2007). Recommendations for
Phys 38:4824–4845. Available from: http://www. radioembolization of hepatic malignancies
aapm.org/pubs/reports/RPT_144.pdf [Accessed using yttrium-90 microsphere brachytherapy:
October 17, 2011]. A consensus panel report from the radioem-
Dieudonné, A. et al. (2011). Clinical feasibility of bolization brachytherapy oncology consor-
fast 3-dimentional dosimetry of the liver for tium. Int J Radiat Oncol Biol Phys 68:13–23.
treatment planning of hepatocellular carci- Lam, M.G.E.H. et al. (2014). Limitations of body
noma. J Nucl Med 52:1930–1937. surface area-based activity calculation for
Eckerman, K.F., Endo, A. (2008). MIRD: radioembolization of hepatic metastases
Radionuclide Data and Decay Schemes, Second in colorectal cancer. J Vasc Interv Radiol
Edition. Reston, VA: The Society of Nuclear 25:1085–1093.
Medicine and Molecular Imaging. Lambert, B.L. et al. (2011). Urinary excretion of
Emami,B. et al. (1991). Tolerance of normal tissue yttrium-90 following intraarterial microsphere
to therapeutic irradiation. Int J Radiat Oncol treatment for liver tumours. J Nucl Med
Biol Phys 21:109–122. 52(suppl 1):1744.
Erwin, W. et al. (2012). A measured bremsstrah- Metyko, J., Erwin, W., Poston, J. Jr, Jimenez, S.
lung dose rate constant for yttrium-90 micro- (2014). 90Sr content in 90Y-labeled SIR-Spheres
sphere liver directed brachytherapy. J Nucl and Zevalin. Health Phys 107:S177–S180.
Med 53(Suppl 1):2230. Metyko, J. et al. (2012). Long-lived impurities
Erwin, W. (2012). Accuracy of yttrium-90 micro- of 90Y-labeled microspheres, TheraSphere
spheres delivery vial exposure rate measure- and SIR-Spheres, and the impact on patient
ment: Effect on estimated percent of activity dose and waste management. Health Phys
delivered. J Nucl Med 53(Suppl 1):2244. 103(5S):S204–S208.
References 165

Mo, L. et al. (2005). Development of activity Ostrowski, R. et al. (2007). Radionuclidic purity
standard for 90Y microspheres. Appl Radiat assessment of yttrium-90 (Y-90) microspheres.
Isot 63:193–199. J Nucl Med 48(Suppl 2):2206.
NCRP. (2006). Report 155: Management of Pasciak, A.S., Erwin, W.D. (2009). Effect of voxel
Radionuclide Therapy Patients. Bethesda, MD: size and computation method on Tc-99m
National Council on Radiation Protection and MAA SPECT/CT-based dose estimation for
Measurements. Y-90 microsphere therapy. IEEE Trans Med
NCRP. (2010). Report 161: Management of Imag 28:1754–1758.
Persons Contaminated with Radionuclides: Petitguillaume, A. et al. (2014). Three-dimensional
Handbook. Bethesda, MD: National Council personalized Monte Carlo dosimetry in 90Y
on Radiation Protection and Measurements. resin microspheres therapy of hepatic metas-
Nelson, K., Vause, P.E., Jr., Koropova, P. (2008). tases: Nontumoral liver and lungs radiation
Post-mortem considerations of yttrium-90 protection considerations and treatment plan-
(90Y) microsphere therapy procedures. Health ning optimization. J Nucl Med 55:405–413.
Phys 95(Suppl 5):S156–S161. Salem, R., Thurston, K.G. (2006).
NRC. (2007). Yttrium-90 TheraSphere and Radioembolization with 90Yttrium micro-
SIR-Spheres Impurities: NRC Information spheres: A state-of-the-art brachytherapy
Notice 2007–10. U.S. Nuclear Regulatory treatment for primary and secondary liver
Commission. Washington, DC. Available malignancies. J Vasc Interv Radiol 17:1251–1278.
from: http://www.nrc.gov/reading-rm/doc- Siegel, J.A. (2004). Guide for Diagnostic Nuclear
collections/gen-comm/info-notices/2007/ Medicine and Radiopharmaceutical Therapy.
in200710.pdf [Accessed October 7, 2011]. Reston, VA: The Society of Nuclear Medicine
NRC. (2008). Consolidated Guidance About and Molecular Imaging.
Material Licenses: NUREG-1556 Vol. 9 Rev. Selwyn, R.G. et al. (2007). A new internal pair
2. U.S. Nuclear Regulatory Commission. production branching ratio of 90Y: the devel-
Washington, DC: Office of Federal and State opment of a non-destructive assay for 90Y and
Materials and Environmental Management 90Sr. Appl Radiat Isot 65:318–327.

Programs. Available from: http://pbadupws. Selwyn, R. et al. (2008). Technical note: The
nrc.gov/docs/ML0734/ML073400289.pdf calibration of 90Y-labeled SIR-Spheres® using
[Accessed March 21, 2011]. a nondestructive spectroscopic assay. Med
NRC. (2012). Microsphere Brachytherapy Phys 35:1278–1279.
Sources and Devices: Licensing Guidance – Simon, B.A. (2000). Non-invasive imaging of
TheraSphere® and SIR-Spheres® Yttrium-90 regional lung function using x-ray com-
ZMicrospheres. Washington, DC: U.S. Nuclear puted tomography. J Clin Monit Comput
Regulatory Commission. Available from: 16:433–442.
http://pbadupws.nrc.gov/docs/ML1217/ Sirtex Medical Limited. (2003). SIR-Spheres®
ML12179A353.pdf [Accessed January 14, Training Program – Physicians and Institutions
2013]. [Accessed June 30, 2010].
NRC. (2013). 10 Code of Federal Regulations Part Sirtex Medical Limited. (2010). Package Insert
35. Washington, DC: U.S. Nuclear Regulatory – SIR-Spheres® Microspheres (Yttrium-90
Commission. Available from: http://www.nrc. Microspheres). Available from: http://www.sir-
gov/reading-rm/doc-collections/cfr/part035/ tex.com/media/29845/ssl-us-10.pdf [Accessed
[Accessed April 25, 2016]. August, 2015].
NRC. (2015). Specialty Board(s) Certification Thieme, K., Beinlich, U., Fritz, E. (2004).
Recognized by NRC under 10 CFR Part Transfer standard for beta decay radio-
35. U.S. Nuclear Regulatory Commission. nuclides in radiotherapy. Appl Radiat Isot
Washington, DC. Available from: http://www. 60:519–522.
nrc.gov/materials/miau/med-use-toolkit/ Zanzonico, P.B., Binkert, B.L., Goldsmith, S.J.
spec-board-cert.html [Accessed March 31, (1999). Bremsstrahlung radiation expo-
2016]. sure from pure β-ray emitters. J Nucl Med
40:1024–1028.
8
The radiation biology
of radioembolization

MARTA CREMONESI, FRANCESCA BOTTA, MAHILA FERRARI, LIDIA


STRIGARI, GUIDO BONOMO, FRANCO ORSI, AND ROBERTO ORECCHIA

8.1 Introduction 167 8.3.3Tumor control probability (TCP) 174


8.2 Basic aspects of radiation biology 169 8.3.4Normal tissue complication
8.2.1 Repair 169 probability 176
8.2.2 Repopulation 169 8.4 Application of the models and examples 180
8.2.3 Reoxygenation 169 8.4.1 NTCP predictions under the
8.2.4 Redistribution 170 hypothesis of uniform irradiation 188
8.2.5 Further considerations specific 8.4.2 NTCP predictions for clinical cases
to the 4Rs in radioembolization 170 with nonuniform irradiation 190
8.3 Radiobiological models 171 8.5 Summary for clinical applications 193
8.3.1 The linear quadratic model 171 8.6 Conclusions 195
8.3.2 The biological effective dose 173 References 195

the treatment of differentiated thyroid cancer and


8.1 INtrODUCtION benign thyroid disease with 131I, neuroblastomas
with 131I-mIBG (metaiodobenzylguanidine), neuro-
In recent years, the use of dosimetry to support endocrine tumors with 177Lu and 90Y radiopeptides,
radionuclide therapy has gained importance as bone pain palliation with 153Sm-ethylene diamine
documented by the increased number of arti- tetramethylene phosphonate (153Sm-EDTM), and
cles addressing dosimetry. The latest literature also radioembolization of primary and secondary
reports  some remarkable correlations between liver cancer with 90Y-microspheres.
absorbed dose delivered, response, and toxic- In particular for radioembolization, significant
ity, which have advanced the understanding of dose–effect correlations have been provided in the
radiobiological effects. In a recent review pub- literature, including a study by Strigari et al. (2010),
lished by Strigari et al. (2011), dose–effect rela- which was the first to describe a model to inter-
tionships were collected and these indicated that pret toxicity and tumor response for hepatocellu-
dosimetry-based personalized treatments would lar carcinoma (HCC) treated with 90Y-labeled resin
improve outcomes and increase survival and open spheres; studies by Garin et al. (2012, 2015, 2016),
the way toward predictivity and personalization which showed prognostication of tumor response
of therapy. Available evidence covers nearly all and survival for HCC treated with 90Y-labeled
widely used nuclear medicine therapies, including glass microspheres; a study by Chiesa et al. (2015),

167
168 The radiation biology of radioembolization

showing dose thresholds for tumor response and dose rate, dose distribution, organ structure, volume
liver toxicity for HCC treated with 90Y-labeled effect, radiosensitivity, combined therapies, and risk
glass microspheres; and finally a study by Flamen factors all at the same mean dose. In the clinical
et al. (2008), showing the prediction of metabolic context of radioembolization, outcomes such as the
response evaluated by multimodality imaging for volume effect, the influence of the functional reserve
metastatic liver tumors and 90Y-labeled resin micro- and/or concomitant therapies, and higher tolerability
spheres. Interestingly, besides providing correla- of retreatment have been empirically observed. This
tion between dose and effect, these investigations reflects a further similarity with EBRT, although dif-
pointed out apparent and unexpected differences ferences with EBRT exist and must be taken in mind.
in tolerability and response associated with glass Moreover, three-dimensional (3D) voxel dosimetry
vs. resin 90Y microspheres, which could not be methods at both a macroscopic and microscopic
resolved by invoking the mere concept of absorbed levels recently applied to radioembolization have
dose, at least if only the mean absorbed dose at provided dose distribution maps and dose–volume
macroscopic level was considered. histograms (DVH) that permit the development of
As a whole, the findings up to now (circa 2016) more refined radiobiological models.
highlight the need for more refined models to This chapter presents the basic aspects of radia-
improve dosimetry information, and especially, tion biology, developed for EBRT and subsequently
the importance of radiation biology to define the adapted to nuclear medicine therapy. The most
effect of radioembolization on tissue. widely used radiobiological models will be dis-
As a natural consequence, the first attempts at cussed, including the linear quadratic model with
defining radioembolization dose–effect relation- the biological effective dose (BED) concept, tumor
ships have been extensively compared with the control probability (TCP), and the normal tissue
data from external beam radiotherapy (EBRT). Of complication probability (NTCP) models. These are
course, in EBRT, the use of radiobiological models is the first of many acronyms common in radiation
well established and enables the possibility of com- biology which will be introduced in this chapter. A
paring effects due to different irradiation modalities, summary of acronyms is provided in Table 8.1.

Table 8.1 Abbreviations and acronyms


BED = biological effective dose N0 = initial number of clonogenic cells
BED50 = BED value for 50% complication NTCP = normal tissue complication probability
probability QUANTEC = Quantitative Analyses of Normal
BEDEBRT, BEDRE = BED in EBRT and in RE Tissue Effects in the Clinic
treatments RE = radioembolization
CTCAE = Common Terminology Criteria for RECIST = Response Evaluation Criteria in Solid
Adverse Events Tumors
D = absorbed dose SF = surviving fraction of irradiated cells
d = absorbed dose delivered in one EBRT T = total duration of exposure
fraction T* = effective time reached when BED = 0
DVH = dose volume histograms TD5, TD50 = tolerated absorbed dose for 5% or
EASL = European Association for the Study of 50% complication probability (at 5 years if
the Liver specified by TD5,5, TD50,5)
EBRT = external beam radiotherapy Tav = doubling time of proliferation
Erel = relative effectiveness (Erel = BED/D) TCP = tumor control probability
EUD = equivalent effective dose Teff = effective half-life
EUBED = equivalent uniform biological effective Tphys = physical half-life of 90Y
dose Trep = repair half-life
fr = fraction of EBRT V = tumor volume
FSU = functional subunits Veff = effective volume
HCC = hepatocellular carcinoma λeff = effective decay constant
LKB model = Lyman–Kutcher–Burman model λphys = physical decay constant
msA = microsphere-specific activity μrep = repair constant = 0.693/Trep
8.2 Basic aspects of radiation biology / 8.2.3 Reoxygenation 169

Finally, some explanatory examples on how as an exponentially decaying function over time,
radiation biology can guide radioembolization with half-time (Trep) varying from minutes to few
planning are also given; the most relevant studies hours.
on these issues are briefly reported as well.

8.2.2 REPOPULATION
Both tumor and normal tissues have a char-
8.2 BaSIC aSPECtS OF
acteristic proliferation rate that allows tumor
raDIatION BIOLOGY
growth and regeneration of some normal tissues
(e.g., the bone marrow and the liver). During
Radiation biology is the study of the effect of irradiation, repopulation counteracts the cell
ionizing radiation on biological tissues. These killing induced by radiation, so repopulation is
complex effects involve physics, chemistry, and desirable following irradiation of normal tissues
biology concepts. The most important biological to limit side effects, while it is unwanted in the
factors that play a role during irradiation, affect- case of tumors due to the potential impairment
ing to some extent the outcome of the treatment, of treatment.
are summarized by the so-called “4Rs of radia- Keeping in mind that radiation-induced cell
tion biology”: repair of DNA damage, redistri- killing includes the loss of the reproductive capa-
bution of cells in the cell cycle (spanning a few bility of the cell, it follows that damage becomes
hours), repopulation (spanning 5–7 weeks), and visible when the cell reaches the phase of mitosis:
reoxygenation of hypoxic tumor areas (span- at that stage, if lethal DNA damage has occurred,
ning a few hours to few days) Pajonk et al. (2010). cell replication is prevented. Therefore, radiation
Certainly tumor response is modulated by response, namely tumor control or organ failure,
many additional factors, and some authors add arises after a latency time that is linked to the pro-
the intrinsic radiosensitivity of individual can- liferation rate in that tissue.
cer stem cells as the fifth R, although this may In the case of tumors, fast growing tumors may
vary during radiation therapy and needs deeper show a decrease in their growth rate within a few
investigation. days after irradiation, while more indolent tumors
may need weeks or months (HCC) to reduce in
size, but still have the possibility to respond com-
8.2.1 REPAIR pletely to therapy (Withers et  al., 1988; Withers,
Ionizing radiation cell killing is a consequence of 1992). Similarly, quickly proliferating tissues such
unrepairable DNA double-strand breaks. Most as bone marrow, skin, and intestinal mucosa may
radiation-induced DNA injury is, however, sub- give a warning sign of damage very early after the
lethal and may be repaired depending on fac- beginning of the treatment. Conversely, slowly
tors including the type and energy of radiation, proliferating tissues such as kidney, liver, lung,
the dose rate, and the phase in the cell cycle. If and bone may manifest injury after months or
radiation is delivered with a low dose rate, the years.
repair possibility increases, while at increasing
dose rates, sublethal lesions can rapidly accumu- 8.2.3 REOXYGENATION
late without full repair, contributing to lethality.
Based on the different ability of normal tissues and The oxygen enhancement ratio (OER) refers to
tumors to repair radiation damage, therapy frac- the enhancement of a therapeutic or detrimental
tionation is a recurrent strategy in EBRT to spare effect of ionizing radiation due to the presence of
normal tissues. oxygen, and is quantitatively defined as the ratio
This concept has also been extrapolated to some between the absorbed dose in hypoxic conditions
nuclear medicine therapies, including radioembo- and normal conditions for a same biological effect.
lization (Cremonesi et al., 2008), dividing the treat- Oxygen is a potent modifier of radiosensitivity and
ment in cycles to reduce toxicity to late responding hypoxic cells are typically two to three times more
organs at risk. The repair probability is described resistant to radiation. Tumors typically contain
170 The radiation biology of radioembolization

regions of transient acute and/or chronic hypoxia, 8.2.5 FURTHER CONSIDERATIONS


especially in the center due to vascularization. These SPECIFIC TO THE 4Rs IN
tumors have often been shown to be associated with RADIOEMBOLIZATION
a poor prognosis. Moreover, there is evidence that
the duration of hypoxic conditions and the extent Radiation therapy using EBRT and radioemboli-
of the hypoxia are influential factors. Although the zation differ in various aspects. First, in EBRT the
underlying mechanisms are to be further clarified, treatment is generally delivered by fractionating
cells irradiated shortly after reoxygenation or after the total dose, e.g., at 2 Gy per fraction (fr), while
long-term exposure to hypoxia are more radiosen- in radioembolization treatment is delivered in a
sitive compared with those irradiated after 4–24 single session and the dose rate decreases over time
hours of hypoxia. due to the physical decay of 90Y. Furthermore, resin
Reoxygenation between dose fractions is microspheres produce an additional embolic effect
generally believed to ease the sterilization of that may contribute to tumor control.
hypoxic cells by increasing tumor radiosensitiv- In particular, the use of fractionation in EBRT
ity. Reoxygenation mechanisms span a few hours permits the redistribution of cells in the hypoxic
to few days and typically occur a few days after the and oxic compartment, which generally occurs in
beginning of irradiation, when the depopulation of the interval between fractions. This phenomenon
the more radiosensitive cells from the bulk tumor arises during dose delivery in radionuclide ther-
enables more hypoxic cells to reach the blood ves- apy but in the case of HCC, cell loss could require
sels, stimulating oxygenation. treatments separated by months as reported in the
clinical report assessing tumor response (Kong
and Hong, 2015). The possibility to increase the
8.2.4 REDISTRIBUTION oxic levels of tumoral cells present in the hypoxic
area before therapy could be possible during radio-
Cells exhibit a different radiosensitivity at differ- nuclide therapy.
ent phases of the cell cycle, with cells in the early From another point of view, large hypoxic areas
S- and late-G2/M phase being most sensitive to contribute to the stability of tumor volume over
ionizing radiation, while cells in late S-phase are time. Stability is considered as a form of tumor
the most resistant. During fractionated radiation control for liver cancer. Moreover, some authors
therapy, cells in the G2/M-phase are preferentially report that chronic hypoxic areas are more radio-
killed, leading to a block of cells in the G2 phase sensitive than oxic ones, while the contrary is true
and to a resulting synchronization of cells in the for acute hypoxic areas. In general, from the radio-
radioresistant S-phase. The time interval between biological point of view, a tumor can be consid-
fractions allows resistant cells from the S-phase of ered to be constituted of both chronic and acute
the cell cycle to unsynchronize, redistributing into hypoxic cells as reported in Strigari et al. (2010).
phases in which cells of both tumors and normal This means that chronic/acute hypoxic areas could
tissues are more radiosensitive, thereby increas- be damaged by radiation and further that dam-
ing the radiation damage. Redistribution effects age may be only partially repaired due to the local
span several hours and play an important role absence of the oxygen.
in EBRT, particularly when fractions are spaced A positive oxygen effect could occur in radio-
out by several hours. In fact, redistribution dur- embolization strategies that plan more than one
ing fractionated irradiation allows the sparing of cycle. The first cycle of radioembolization should
normal tissues that have few rapidly cycling cells provoke a partial tumor shrinkage, potentially
compared with tumors containing many cells with increasing the vascular perfusion to the remaining
rapid turnover. tumor. This should facilitate oxygenation and thus
The effects summarized by the 4Rs have a lead- increase radiosensitivity, enhancing the effects of a
ing role for the success and optimization of radia- subsequent cycle. Such an approach, although clin-
tion therapy, including radionuclide therapy, and ically more complex, could improve the response
have represented a landmark for the development of tumors, compared with a single radioemboliza-
of radiobiological models describing the survival tion therapy. Increased efficacy is likely, especially
of cells after irradiation. in the case of tumors large in size and with areas
8.3 Radiobiological models / 8.3.1 The linear quadratic model 171

poorly vascularized but not necrotic, or in cases acidosis, oxidative stress, and nutrient depriva-
where potential toxicity to normal tissues is of tion (Mitsuishi et al., 2012). Unfortunately, these
concern. factors are only partially included in the radio-
Furthermore, radiotherapy using both exter- biological models. Additional studies are needed
nal beam and internal emitters is a therapeu- to further address these issues.
tic strategy based on the oxidative stress. Both
treatment modalities are specifically designed
to increase reactive oxygen levels in tumor cells
to elicit their death through sudden and intense 8.3 raDIOBIOLOGICaL MODELS
oxidative stress (Manda et al., 2015). Cancer cells
present a high intrinsic oxidative activity, so less 8.3.1 THE LINEAR QUADRATIC
additional reactive oxygen species are required MODEL
compared with normal cells for triggering cell
death. Levels of reactive oxygen species that Among radiobiological models describing the sur-
are cytotoxic for cancer cells induce less drastic vival of cells after irradiation, the linear quadratic
effects in normal cells, which have a lower oxida- model is the most well known.
tive status and are endowed with efficient systems A cell survival curve describes the relationship
to repair injuries induced by reactive oxygen between the fraction of surviving cells (SF), i.e.,
species—within certain limits. Nevertheless, the fraction of irradiated cells that maintain repro-
precise targeting of dose to the diseased tissue is ductive integrity, and the absorbed dose. Its shape
a priority, aiming to spare normal tissues against depends on several factors, including the type of
the deleterious action of “therapeutic” reactive radiation, the type of cells, and the radiation dose
oxygen species. The sparing of normal tissue is rate.
guaranteed in EBRT by the possibility of using The linear quadratic model describes the effect
advanced delivery techniques, while in radio- induced by radiation in a cell population as a func-
embolization it is achieved by selective or super- tion of the dose delivered, and from that, the SF.
selective administration of radioactive sources The main hypothesis assumed by the linear qua-
within the liver. dratic model is that the SF of cells receiving an
In addition, the macroembolic effect of resin instantaneous absorbed dose D (as occurs, e.g., in
microspheres is accompanied by a greater lack EBRT) follows the equation:
of oxygen resulting in ischemia, and therefore,
enhanced efficacy. On the other hand, a shortage ln(SF) = (−αD −βD 2 ) (8.1)
of oxygen might also diminish the tumoricidal
effect of ionizing radiation due to a lack of oxy- or, equivalently,
gen radicals. In other words, the embolic effect
SF = exp(−αD −βD 2 ) (8.2)
and the potential reduction of oxygen free radi-
cals are opposite phenomena and the resulting The radiation-induced damage is described by
final net effect of these processes is still unclear. the sum of two terms, αD and βD2, respectively,
The process of tumor control or normal tis- representing
sue damage is complex with respect to the cur-
rently proposed modeling (Strigari et al., 2011; ● DNA irreparable events (double-strand breaks)
Cremonesi et al., 2014). Increased cell dam- in which both strands in the double helix are
age after repair signaling can cause mitotic simultaneously severed. The number of such
catastrophe and cell death with an associated events is proportional to D by the factor α,
inflammatory tissue response. Tissue changes which represents the intrinsic radiosensitiv-
due to significant cell death can alter oxygen- ity. α is tissue specific and describes the initial,
ation status (reoxygenation) and trigger acceler- linear slope of the SF curve.
ated repopulation and redistribution in the cell ● Two independent DNA reparable events
cycle. Finally, tumor heterogeneity derives also (single-strand breaks), occurring close enough in
from the nonuniform spatial distribution of time and space on the DNA filament to gener-
microenvironmental stresses, such as hypoxia, ate cellular death. The number of such events
172 The radiation biology of radioembolization

is proportional to D2 (since it is a combination of α/β values, corresponding to a marked impact of


two independent events, each having an occur- the quadratic term; the opposite occurs for tissues
rence proportional to D) by a factor β, smaller that exhibit high α/β values, with lower relevance of
than α, which represents the potential sparing the quadratic term (−αD − βD2 ≅ −αD). The effect
capacity. β is also tissue specific and describes of dose rate on SF for dose rates typical in common
the quadratic slope of the SF curve. radiation therapy modalities, including 90Y radio-
embolization, is illustrated visually in Figure 8.2.
The ratio α/β represents the dose at which the To account for this in the formalism, the qua-
linear and quadratic components of cell killing are dratic term needs to include an explicit dependence
equal and describe the shape of the SF curve. The on the dose, dose rate, repair half-time Trep, and the
features of the linear quadratic model are described total duration of exposure, T. The Lea–Catcheside
visually in Figure 8.1. factor is a positive, dimensionless function g(T)
Repair mechanisms play a role in the quadratic ranging between 0 and 1 (Millar, 1991; Brenner
component of DNA damage. In fact, a single- et al., 1998; Baechler et al., 2008), which is coupled
strand break that is not repaired can potentially to the quadratic component to express reduced cell
combine with another single-strand break occur- killing due to increased repair of sublethal damage
ring close to it in space and time, and result in during a continuous irradiation and/or between
damage. Conversely, if the first single-strand fractions:
break is repaired before the occurrence of another
single-strand break, no lethal effect will occur. This SF(D ) = e
(−α.D. − g (T )⋅β. D 2 ) (8.3)
is more likely to happen if radiation is delivered
with a low dose rate, which means that there is a In the case of irradiation with a decaying source
longer time interval between any two consecutive having an effective half-life Teff, if the dose delivery
single-strand breaks. Thus, the probability of cell is protracted for a time T significantly longer than
death increases with the increasing dose rate. This the repair half-time (T >> Trep), then g(T) can be
is particularly true for tissue characterized by low approximated by

1.E+00
α radiosensitivity

β sparing capacity
1.E–01
exp(–αD)

exp(–βD2)
SF

exp(–αD – βD2) α⨂β


1.E–02

1.E–03
0 5 10 15 20 25
Dose (Gy)

Figure 8.1 Curves describing the surviving fraction (SF) of cells vs. absorbed dose. The dashed curve
represents the linear component of the SF related to DNA irreparable lesions resulting from a single
hit, with the parameter α associated to the radiosensitivity of the specific tissue. The dotted curve
describes the quadratic component of the SF resulting from two DNA sublethal lesions occurring
close enough in time and space to create lethal damage. β is associated to the sparing capacity for
the specific tissue. The continuous curve represents the SF curve with both α and β components.
8.3 Radiobiological models / 8.3.2 The biological effective dose 173

1.E+00

90
Y
1.E–01

1.E–02 LDR

1.E–03 EBRT
SF

HDR

1.E–04
1 Gy/min

ra ng
se asi
te
do c r e
0.01 Gy/min
1.E–05

in
0.001 Gy/min

1.E–06
0 5 10 15 20 25 30
Dose (Gy)

Figure 8.2 Typical trend for SF of cells following irradiation at different dose rates (dotted curve:
0.001 Gy/min; dashed curve: 0.01 Gy/min; continuous curve: 1 Gy/min). The areas characterized by
dose rate values typically applied for EBRT, high-dose-rate brachytherapy (HDR), low-dose-rate
brachytherapy (LDR), and 90Y radioembolization (90Y) are also indicated.

λ eff Trep SF = exp(α ⋅ BED) (8.6)


g (T >> Trep ) ≈ = (8.4)
λ eff + µ Trep + Teff
By comparison with Equation 8.3, BED can also
where μ = 0.693/Trep and λeff = 0.693/Teff (Strigari be defined as the product of D and a modifying fac-
et al., 2011). tor (named relative effectiveness, Erel) that includes
Typically, α/β is assumed to be 2.5 Gy for nor- dose rate effects depending on radiosensitivity and
mal tissues and 10 Gy for tumors. For radioembo- repair of damage:
lization, Teff equals the physical half-life of 90Y (Tphys
= 64.24 hours, i.e., λeff = λphys  =  0.0108  hour –1), BED = D·Erel = D(1 + g(T)·D·β/α) (8.7)
while Trep is equal to 2.5 hours (μrep = 0.28 hour –1) In the case of EBRT, with a total dose DEBRT
for normal liver and 1.5 hours (μrep = 0.53 hour –1) delivered in N equal fractions (di,EBRT = DEBRT/N),
for tumor (Cremonesi et al., 2008, 2014). A con- from the comparison of Equations 8.1 and 8.5 it
tinuous low dose rate, such as is the case in follows that the BED for each fraction, BEDi, EBRT, is
radioembolization, confers the same benefits as
α ⋅ BEDi ,EBRT = α ⋅ di ,EBRT +β⋅ di ,EBRT2 (8.8)
fractionation, sparing the normal tissues—with
lower α/β value—more than the tumor character- For the whole treatment, the total SF will be the
ized by higher α/β value. product of the single fractions SFi, so
α·BEDEBRT = Σi (α·di,EBRT + β·di,EBRT2) = α·N·di,EBRT
8.3.2 THE BIOLOGICAL EFFECTIVE + β·N·di,EBRT2 = α·DEBRT + β·DEBRT·(DEBRT/N)
DOSE and
 D /N 
The biological effective dose (BED) is defined by ( Erel )EBRT = 1 + EBRT  (8.9)
the natural logarithm of the surviving fraction  α/β 
function versus absorbed dose as in the following In the case of radioembolization, with a total
expression: dose delivered, DRE:
ln(SF) = −α ⋅ BED α ⋅ BEDRE = α ⋅ DRE + g (T ) ⋅β⋅ DRE 2
(8.5) (8.10)
174 The radiation biology of radioembolization

and including the expression of g(T) from Equation The following equations relate EQ to BEDEBRT and
8.4: BEDRE:
 DRE ⋅ λ eff  
( Erel )RE = DRE ⋅ 1 +  (8.11) d 
BEDEBRT = EQ 1 +  = BEDRE (8.16)
 (µ rep + λ eff ) ⋅ α / β   α/β
or thus
 DRE ⋅ Teff  BEDRE ⋅ α / β
( Erel )RE = DRE ⋅ 1 +  (8.12) EQ = (8.17)
 (Trep + Teff ) ⋅ α / β  d +α /β

For tumor tissue, an additional term can be added


to the BED expression accounting for repopulation. 8.3.3 TUMOR CONTROL
This phenomenon competes with cell killing, so it PROBABILITY (TCP)
appears as a term, the biological repopulation factor
(BRF), which reduces the effect of irradiation: Considering a tumor uniformly irradiated with a
certain BED, the function that describes the tumor
BED = DErel − BRF (8.13)
control probability is defined (Strigari et al., 2011) by
BRF is the biological equivalent of tumor repop-
ulation that occurs during treatment and which TCP(BED) = exp[− N 0 ⋅ SF(BED)]
(8.18)
effectively “wastes” some of the delivered dose = exp[− N 0 ⋅ exp(−α ⋅ BED)]
(Dale, 1996; Dale et al., 2000). BRF increases with
the duration of the treatment, T, during which the where N0 is the initial number of clonogenic cells
repopulation occurs. Considering exponential cell and α is the radiosensitivity parameter previously
proliferation with doubling time Tav, Equation 8.10 defined. N0 is the typically proportional to the
becomes (Dale, 1996; Antipas et al., 2001) tumor volume V, so N0 = ρV is the strongly intra-
tumor and intertumor dependent (ρ is the density
ln2
BED = DErel − KT = DErel − T (8.14) number of clonogenic cells) (Wigg, 2001).
αTav The mathematical formulation expressed by
For EBRT the whole duration of treatment, T, is Equation 8.18 is valid under the assumption that
the overall treatment time. radiation-induced cell killing is ruled by Poisson sta-
In radionuclide therapy, with continuously tistics. Poisson statistics describe the observations of
decaying dose rate, the treatment is assumed to random events, each having a small probability of
be concluded after a so-called “effective time,” T*, success, for which the average number of successes
which is reached when the BED of Equation 8.10 (Ϛ) after an “experiment” involving a large number
equals zero. After the effective time, the repopula- of events has been defined. Also, the events must be
tion exceeds the cell killing and the dose delivered independent, namely, the outcome of one event does
is considered to be wasted. not affect the outcome of the others. Under these
It can be demonstrated (Antipas et al., 2001) that assumptions, the probability of observing ϕ success-
 ln2  ful events after a large number of events is given by
1 (8.15)
T* = − ⋅ ln  
λ eff  α ⋅ R0Tav  e−ς ⋅ς ϕ
P(ϕ ) = (8.19)
ϕ!
The BED is widely used to compare different In our case, the “experiment” is represented by
radiation therapy modalities, for example, EBRT the irradiation of tumor cells, the (large) number
delivered with different fractionation schemes or of events is quantified by the absorbed dose D or
EBRT versus radionuclide therapy. Two different BED. The “successful” event is represented by a
radiation modalities delivering the same BED are single cell surviving after being hit by radiation,
expected (by definition) to produce the same effect. and the average number of “successful events” after
The equivalent dose (EQ) is the absorbed dose the “experiment,” Ϛ, is given by the average num-
delivered with EBRT in N fractions of dose d each ber of cells surviving after irradiation:
(EQd = Nd), leading to a BED (BEDEBRT) that is the
same as a radioembolization treatment (BEDRE). ς = N 0 ⋅ SF(BED) = N 0 ⋅ exp(−α ⋅ BED)) (8.20)
8.3 Radiobiological models / 8.3.3 Tumor control probability (TCP) 175

The hypothesis of event independence is repre- After dividing the possible BED range into a
sented by the assumption that the death of a cell number M of intervals, Ni represents the number
due to radiation does not affect the successive of voxels receiving a BED value included in the ith
probability of cell killing in any other cell. interval and BEDi is the average BED value in that
TCP is represented by the probability of having same interval. With N defined as the total num-
no surviving cells, which, under the previous for- ber of voxels constituting the tumor, the EUBED
malism, is the probability of having no “successful” becomes
events. According to Equation 8.19 TCP becomes
1 SFi 
M

TCP = P(0) = e−ς (8.21) EUBED = −  ln


α

∑i
N 


which is equivalent to Equation 8.18 after combin- (8.24)
1 exp(−α ⋅ BEDi ) 
M
ing Equations 8.20 and 8.21.
In the case of nonuniform irradiation of the = −  ln
α

∑ N


tumor, the same formalism in Equation 8.18 i

can be maintained after calculation involving


In the extreme case of infinitely small intervals,
the absorbed dose (or BED) distribution over
each BED value represents an interval itself and
the tumor volume, which must be known. If we
Equation 8.24 becomes
assume that the BED distribution in the tumor
volume is described by a probability function,
1 exp(−α ⋅ BEDi ) 
N
P(BED), then for each BED value, P(BED) indi-
cates the fraction of the total tumor volume
EUBED = −  ln
α

∑ i
N
 (8.25)

receiving a biological effective dose equal to
BED. The SF value over the entire tumor volume In this case, the TCP is calculated by
is obtained by calculating the surviving fraction TCP({N i ,BEDi }) = TCP(EUBED)
associated with each BED value (SF(BED) = exp(− (8.26)
αBED)) and by opportunely weighting each value = exp(− N 0 ⋅ exp(−α⋅ EUBED))
according to its probability of occurrence:
Examples of application of the TCP concept to

radioembolization clinical data can be found in

SF = P(BED) ⋅ e −α⋅BED d BED
0
(8.22) Strigari et al. (2010) and Chiesa et al. (2015), for
the treatment with resin and glass microspheres,
A new parameter has been defined, the equiva- respectively.
lent uniform biological effective dose (EUBED) Strigari et al. (2010) defined the tumor response
(O’Donoghue, 1999; Jones and Hoban, 2000), rep- according to the Response Evaluation Criteria in
resenting the BED, which, if given uniformly over Solid Tumors (RECIST) and European Association
the whole tumor volume would produce the same for the Study of the Liver (EASL) criteria account-
fraction of surviving cells as the nonuniform BED ing for tumor necrosis when evidenced by non-
distribution described by P(BED) in Equation 8.22. enhanced areas. Equation 8.18 was adopted,
Following Equation 8.5 including a further term able to take into account
possible variation in the density of clonogenic cells
1 1 in the patient’s tumor:
EUBED = − ln(SF) = −
α α
(8.23)  

ln( ∫ P(BED) ⋅ e−α⋅BEDdBED)
0
TCP(D ) = exp  −

∑i
ηi ⋅ N 0 ⋅ SF(BED) (8.27)

In clinical practice, the BED distribution is not where


expressed by a continuous function P(BED), but a
discrete formulation {Ni, BEDi}, which can be eas- 1 1  1  ln( N ) − ln( N )  2 
ily obtained if the 3D absorbed dose distribution is ηi = ⋅ ⋅ exp −  i 0
  (8.28)
2π σ ln( N ) 2  σ
available and converted into a 3D BED map.  ln( N )

176 The radiation biology of radioembolization

ηi is the proportion of population having a dose distribution at the microscopic level, which
number of clonogenic cells equal to Ni, derived necessitates higher average doses to reach an effect
from a Gaussian distribution of ln(N) with a mean comparable with EBRT. Chiesa et al. (2015) also
value of ln(N0) and a standard deviation σln(N). The investigated the possible variation of TCP parame-
fit of experimental data indicated the presence of ters with tumor size, dividing the whole pool of data
two subpopulations, a more radioresistant one in two groups. Substantially different trends were
with α  = 0.001 Gy –1 and ln(N0) = 23, and a less observed: the absorbed dose yielding a 50% TCP was
radioresistant one with α = 0.005 Gy –1 and ln(N0) around 250 Gy for lesions smaller than 10 cm3, while
= 6.9 (Strigari et al., 2010). According to both cri- 50% TCP was achieved at 1300 Gy in large volumes.
teria (RECIST and EASL), an average absorbed
dose to the tumor higher than 110–120 Gy guar- 8.3.4 NORMAL TISSUE
anteed partial or complete response to 50% of the COMPLICATION PROBABILITY
patients. However, following a more detailed anal-
ysis, the two criteria for tumor response produced The literature offers several NTCP models, either
different results in some cases, and the authors developed to fit the phenomenological curves from
suggested a combination of both with the further EBRT and then extended/adapted to other treatment
inclusion of additional criteria based on 18-fluoro- modalities or based directly on concepts derived
2-deoxyglucose positron emission tomography/ from radiation biology, physics, and physiology.
computed tomography (18F-FDG-PET) imaging. The modeling of NTCP for the liver is of spe-
The authors did not account for the heterogeneity cial interest in the context of radioembolization
of dose distribution in their analysis, but remarked since irradiation of normal liver can become a
the need to adopt a more advanced mathematical limiting factor for therapy. Three main models
formalism to consider this issue as well. have been used to interpret dose–effect correla-
Chiesa et al. (2015) also classified the tumor tions for the liver: the Lyman–Kutcher–Burman
response according to the radiological EASL cri- (LKB) model, derived from EBRT (Lyman, 1985;
teria. CT scans were performed every third month Kutcher and Burman, 1989); the parallel architec-
and the best tumor response over time was con- ture model (Withers et al., 1988; Yorke et al., 1992;
sidered. Tumor control was defined considering Jackson et al., 1993; Niemierko and Goitein, 1993);
both complete responding and partial respond- and the model by Walrand, based on dosimetric
ing lesions. TCP data were plotted as a function simulations at the microscopic level (Walrand et
of the average absorbed dose to the lesion, and the al., 2014a, 2014b) for two types of microspheres
parameters α, N0, and g(T)·β were obtained from providing different microsphere-specific activ-
the fit of experimental data with two simplified ity (msA) of 0.05 and 2.5 kBq per microsphere for
expressions of Equation 8.26: resin and glass microspheres, respectively.
Here we present the LKB and the parallel
TCP = exp(− N 0 ⋅ exp(−α ⋅ D − g (T ) ⋅β⋅ D 2 )) (8.29) architecture models. Please refer to Chapter 9,
Section 9.12, for a deep description of the model
or neglecting the quadratic term (i.e., in the by Walrand.
assumption of negligible influence of the dose-rate
effect): 8.3.4.1 The Lyman (and LKB) model
TCP = exp(− N 0 ⋅ exp(−α ⋅ D )) (8.30) The first formulation of the Lyman model (Lyman,
1985) was developed to predict the effect after uni-
The values obtained for both N0 and α were much form EBRT irradiation of whole organs. Clinical data
lower than the corresponding values reported for were collected with reference to a specific endpoint
EBRT: α = 0.002 Gy –1 versus the typical EBRT value (i.e., complication) and the following expression was
of around 0.01 Gy –1, and N0 ranging from 2.7 to 3.4. proposed to fit the absorbed dose–response data:
The apparent lower radiosensitivity exhibited fol-
t
lowing radioembolization should be interpreted—  x2 
according to the authors—as a consequence of
the high heterogeneity of microspheres and hence
NTCP (D ) =
1
2π ∫
−∞
exp −  dx
 2
(8.31)
8.3 Radiobiological models / 8.3.4 Normal tissue complication probability 177

with Using the effective volume method, NTCP can


D − TD50 be calculated using Equation 8.31 assuming that a
t= (8.32) uniform dose equal to the maximum dose in the
m ⋅ TD50
DVH, Dmax, is delivered to a volume called effective
where D is the uniform absorbed dose in the volume Veff, calculated by
organ (delivered at a specified dose per frac-
tion), TD50 is the absorbed dose value at the same
dose per fraction for which 50% of the popula-
Veff = ∑v ( D / D
i i max )1/n (8.34)
tion exhibited complications at a fixed time NTCP can be calculated by Equations 8.31 and
point after the irradiation (e.g., 5 years), and m 8.32 considering D = Dmax and TD50 (Veff/Vtot) cal-
is a parameter representing the steepness of the culated with Equation 8.33.
dose–effect curve. Alternatively, the nonuniform irradiation {vi,
For each effect (complication) considered, the Di} can be reduced to a whole organ irradiation
TD50,5 and m parameters have been experimen- with uniform dose equal to EUD, calculated by
tally obtained from epidemiological data. Similar
curves can be obtained for different fractionation
schemes, different endpoints, or at different time
EUD = ( ∑v ( D )i i
1/n n
) (8.35)

points, with each scenario described by its own NTCP can be calculated by Equations 8.31 and
TD50 and m parameters. In case of partial irradia- 8.32 considering D = EUD and TD50(1). To adapt
tion of an organ, for example, when only a frac- this model to radioembolization, it would be nec-
tion ν of the whole volume is irradiated, but still essary first to calculate the radioembolization
with uniform absorbed dose in the irradiated area, BED volume histogram, then convert the absorbed
the same formalism can be adopted for NTCP esti- dose values into the equivalent dose of EBRT as
mation (Equation 8.31) provided use of properly described by Equation 8.17. EUD or Veff could then
adapted value of TD50: be computed using the newly obtained DVH and
finally, the NTCP could be estimated considering
TD50 ( ν) = TD50 (1) ⋅ ν−n (8.33)
EUD/Veff and curve parameters (TD50,m), which
where n is a parameter, in case of partial irradia- refer to the same fractionation scheme used for the
tion, that quantifies the behavior of the organ and DVH conversion.
to what extent NTCP is affected by the amount of The very first publication providing values for
the irradiated volume ν. Usually, n is restricted to the parameters of the Lyman model was in 1991
the 0–1 interval, where an n value close to zero by Burman et al. (1991) based on the partial liver
indicates that NTCP presents a weak variation for tolerance data published by Emami et al. (1991).
different irradiated volumes, with the converse Subsequently, more updated sets of parameters
true for n approaching 1. In liver, the values of n were presented by Dawson et al. (2001, 2002) and
found by various authors are quite close to 1 (0.95– Dawson and Ten Haken (2005). More recently,
1.1), indicating a notable volume effect (see Figures radiation liver injury and dose–volume data have
8.4 and 8.6 and Example 8.2). The power law of been reviewed in the framework of the Quantitative
Equation 8.33 is not based on a biological or physi- Analyses of Normal Tissue Effects in the Clinic
ological rationale but is a simple expression that (QUANTEC) project (Pan et al., 2010).
adequately describes clinical observations (Yorke It is important to remember that the model
et al., 2001). has been conceived and developed in the frame-
The Kutcher and Burman reduction scheme work of EBRT, which originally aimed to deliver
(Kutcher and Burman, 1989) extends the Lyman a nearly uniform irradiation to the target, with
model to the case of nonuniform irradiation, with an accepted nonuniformity within a few percent.
fractions of volumes vi uniformly irradiated with The adoption in radioembolization of the same
an absorbed dose Di. In this case, the NTCP for- parameter values (m, TD50) for EBRT should
malism (Equation 8.31) can still be maintained as be avoided, or at least made with caution, even
long as the effective volume method or the equiva- after appropriate conversion of radioemboliza-
lent uniform dose (EUD) concept and DVH reduc- tion absorbed dose into the equivalent EBRT
tion are applied. value. This is because the dose delivery during
178 The radiation biology of radioembolization

radioembolization can be very heterogeneous, as a function of different dosimetric and radio-


as described in Chapter 9. When considering biological quantities, including average absorbed
the healthy parenchyma only (thus excluding dose to the uninvolved liver, average BED, EUD,
the tumor), the LKB model exhibits high sensi- and EUBED; two different sets of radiobiological
tivity to the presence of small high-dose regions parameters (either gathered from EBRT expe-
(Yorke et al., 1999), possibly leading to unrealis- rience or self-derived from analysis of radio-
tic NTCP estimates due to the heterogeneity of embolization patients’ data) and two different
dose distribution in radioembolization. This will methods for volume delineation (SPECT-based
be illustrated by some examples in Section 8.4.5.2 or CT-based) were adopted. For each combina-
(Figures 8.14 and 8.15). tion of these variables, a fit of the experimental
Moreover, the heterogeneity of dose delivery data was performed according to Equation 8.31
in the case of radioembolization may be pres- and the TD50 and m parameters were derived. In
ent on both a microscopic and macroscopic scale all cases, the TD50 values obtained were signifi-
(Walrand et al., 2014b) due to the pattern of cantly higher than those adopted for EBRT. For
microspheres distribution in the arteries; in light example, when considering the absorbed dose,
of this, even for an apparent homogeneous dose TD50(1) ranged from 97 to 106 Gy, much higher
delivery, as shown by macroscale posttreatment than the EBRT values, which are typically lower
imaging, microscopic inhomogeneities will still than 50 Gy (Dawson and Ten Haken, 2005; Pan
create discordance with EBRT. Nonetheless, it is et al., 2010). For radioembolization using glass
possible to adopt the LKB formalism (Equations microspheres, Chiesa recommended an average
8.31 and 8.32) to fit the radioembolization dose- absorbed dose to the normal liver equal to 75 Gy
toxicity data and derive a dedicated set of param- corresponding to an accepted complication prob-
eters suitable to make predictions for patients with ability equal to 15% (TD15) to guide the treatment
comparable clinical characteristics, treatment planning.
modalities and dosimetry protocols.
As for TCP, examples on this can be found in
Strigari et al. (2010) for RE with resin microspheres 8.3.4.2 The parallel architecture
and Chiesa et al. (2015) for glass spheres. model
Strigari et al. (2010) classified toxicity accord-
ing to the Common Terminology Criteria for The architecture model assumes that the organ is
Adverse Events (CTCAE) and all events with organized in multicellular entities, called func-
toxicity grade equal or higher than 2 were rec- tional subunits (FSU), that respond to irradia-
ognized as complications. Average BED to tion according to the cell radiosensitivity and to
the liver was calculated and plotted with the the minimum number of cells that needs to be
observed complication probability associated to alive to keep the FSU functioning (Withers et al.,
each BED value; data fit performed according to 1988).
Equation 8.31 resulted in a BED50 estimate equal The occurrence of complication in the whole
to 93 Gy and m equal to 0.28 (assuming n = 1 in organ after irradiation depends on the radiation
case of partial irradiation, Equation 8.34). This distribution among the different FSUs, the radio-
value for BED50 is higher than the values reported sensitivity of each FSU, and on how the different
for late effect induction following EBRT, which is FSUs collaborate to guarantee organ function.
72 Gy according to Emami et al. (1991) and 64 Two basic architectures are defined to represent
Gy according to Dawson et al. (2002). The distri- the organization of cells, the serial and the parallel
bution of microspheres in the vasculature of the architecture. The serial architecture assumes that
vessels determines the absorption of high doses the different FSUs work in series and the impair-
in small volumes, with a possible loss of biologic ment of even a single subunit is able to compro-
effect. mise the whole organ functionality. Conversely,
Chiesa et al. (2015) reported treatment-related the parallel architecture describes the organ as
liver disease according to international guide- many FSUs working in parallel, each nearly inde-
lines, avoiding those clearly attributable to tumor pendent from the others, and states that the whole
progression. Complication probability was plotted organ functionality is preserved until the fraction
8.3 Radiobiological models / 8.3.4 Normal tissue complication probability 179

of impaired subunits is lower than a certain fixed the model (d50, k, fr50 and σfr) have been obtained
amount, referred to as a “functional reserve.” from the analysis of clinical complication data of
Focusing on the liver, its functionality can be patients undergoing EBRT (Jackson et al., 1995).
described by a parallel architecture model (Jackson The best-fit parameters, together with the 68% con-
et al., 1995), which we will now illustrate. A dose– fidence interval, are fr50 = 0.497 ± 0.043; σ fr = 0.047
response function is defined for each subunit, with ± 0.027; d50 = (41.62 ± 3.5) Gy delivered at 1.5 Gy
p(d) the probability, p, of damaging a subunit after fractions; k = 1.95 ± 0.77.
irradiation at a given dose d. A sigmoidal dose– Yorke et al. (1999) investigated the possibil-
response function has been proposed to describe ity of using this model to explain clinical obser-
the subunit response phenomenologically: vations after liver radioembolization using glass
microspheres, where the absence of complication
1
p(d ) = (8.36) was observed after estimated absorbed doses to
1 + (d 1 / d )k  the normal liver up to 150 Gy. First, to apply the
 2 
model to radioembolization, the d1/2 parameter
where d1/2 represents the dose at which 50% of the reported above must be converted into a BED50
subunits is damaged and k is the slope of the dose– value according to Equations 8.7 and 8.9, and the
response function. radioembolization DVH needs to be converted in a
The fraction of an organ damaged by the treat- BED volume histogram according to Equations 8.7
ment, f, can be calculated from the healthy liver and 8.11. Such BED values should replace the d val-
differential DVH assuming that the organ is ues in Equations 8.36 and 8.37. Yorke et al. (1999)
composed of a large number of subunits which found that the value of Trep strongly influenced the
respond to radiation independently and that each NTCP estimate and that only short repair times
FSU is small enough to consider their individual (≤1 h) allowed consistency between the model and
irradiation to be homogeneous. In addition, when clinical data. Second, it was shown that for a same
the number of subunits is large, the total damage average dose to the whole liver, different DVHs
is demonstrated to be well approximated by the result in radically different NTCP estimations. For
mean damage over the voxels, thus yielding: this reason, the question was raised about which
f= ∑v p(d )
i i (8.37)
DVH is adequate to describe the absorbed dose dis-
tribution during radioembolization, which exhib-
i
its steep absorbed dose gradients that may not be
with vi being the fraction of voxels receiving dose di. described by the 3D dose distribution obtained
To calculate the probability that the damage from posttreatment imaging. In the Yorke study
of a fraction f of the organ actually results in a (Yorke et al., 1999), when the parallel architecture
complication, a comparison is made to the distri- model was applied using DVH derived from auto-
bution of functional reserve in the patient popula- radiography, consistency with clinical data was
tion. Assuming that the cumulative distribution is found for model parameters varying in the range
described by a displaced error function H with a indicated by Jackson et al. (1995). When the par-
mean value of the functional reserve fr50 and width allel architecture model was applied using DVH
σfr, the complication probability (NTCP) associ- derived from a simulation of the microsphere dis-
ated to the damage of a fraction f is given by tribution superimposed to the FSU structure (the
liver lobule), consistency with clinical data was
NTCP = H ( f ) found for a wider range of parameters. The authors
f
concluded that the parallel architecture model
1
=
2 πσ 2
fr

0
d(fr) exp[−(fr − fr50 )2 / 2σ 2fr ] (8.38) could be adequate to describe clinical observations
on hepatic toxicity, but that a deeper investigation
on microsphere distribution and mechanisms of
In other words, when a patient suffers the damage repair was needed.
damage of an organ fraction equal to f, the prob- The dose-toxicity model recently presented
ability of observing a complication is estimated by Walrand (Walrand et al., 2014a, 2014b) and
as the proportion of population having a func- described in Chapter 9 has important commonal-
tional reserve lower than f. The four parameters of ities with the parallel architecture model adapted
180 The radiation biology of radioembolization

to radioembolization by Yorke et al. (1999). These approaches. Considering inherent differences


common features include the concept of the between the two kind of therapies, EBRT mod-
FSU, the use of an EBRT-derived sigmoid dose– els were derived for uniform dose delivery, while
response function for the irradiation of a single radioembolization dose distribution is typically
FSU, the development a microscopic model sim- nonhomogeneous not only at macroscopic level
ulating the FSU structure, and the microsphere (i.e., at the voxel level shown by nuclear medicine
distribution to derive the dose distribution at imaging) but also microscopically. In addition,
the lobule level. Walrand’s model parameters resin and glass microspheres exhibit very differ-
are derived from clinical data, and, notably, also ent patterns of dose distribution at the micro-
incorporate the dependence on the mSA, allow- scopic level due to the differences in the number
ing predictions for both resin and glass radioem- of microspheres. Resin microspheres, with a lower
bolization therapy. activity load, 0.05 kBq/sphere, are much more
numerous and can distribute more regularly in
capillaries, providing an overall increase in dose
uniformity compared with glass microspheres.
8.4 aPPLICatION OF tHE The microdosimetric behavior of glass and resin
MODELS aND EXaMPLES microspheres has been illustrated by Walrand
(Walrand et al., 2014a).
In radioembolization, treatment-planning tech- The use of EUD and EUBED concepts repre-
niques suggested by the manufacturers are based sents a first attempt to account for nonuniformity,
essentially on empiric or, at best, rough dosimetry but they are limited to the macroscopic level. In
algorithms (more details provided in Chapter 5). certain circumstances, only a microscopic model-
In the latter case, threshold dose limits were pro- ing could better describe the results, as discussed
posed to preserve the liver, namely 120 Gy to the further in Chapter 9.
whole liver or the treated lobule according to a Here, follow some examples that show various
monocompartmental model proposed for glass steps of the comparison between RE and EBRT,
microspheres, and 80 Gy to normal liver, accord- allowing use of EBRT evidence as a first guide for
ing to a multicompartmental model proposed for the interpretation of the outcomes observed in RE.
resin microspheres. Such threshold values were Unless otherwise specified, all of the examples
included in the clinical trials for both radioembo- provided here consider the following set of radiobio-
lization products and are still routinely used. logical parameters taken from the literature (Dawson
Other researchers preferred to embrace a cau- and Ten Haken, 2005; Cremonesi et al., 2008):
tionary approach and have performed feasibil-
ity studies with escalating dose (Cremonesi et al., 1. α/β = 2 Gy; T1/2 rep = 2.5 hours; T1/2 eff = T1/2 phys
2008; Sangro et al., 2008). The basic idea was to Y-90 = 64.2 hours, for normal liver.
extrapolate constraints from EBRT and establish 2. α/β = 10 Gy; T1/2 rep = 1.5 hours; T1/2 eff = T1/2 phys
an absorbed dose limit to the healthy liver. A limit Y-90 = 64.2 hours, and Tav = 30 days for tumors.
of 40 Gy in radioembolization for a whole-liver
treatment was initially identified as a good com- Figure 8.3a illustrates the BED curve as a func-
promise (see Example 8.1). The first clinical cases tion of the absorbed dose for radioembolization
of liver toxicity manifested at doses higher than and EBRT for the uninvolved liver considering 1.5
40–50 Gy for whole-liver treatments with resin Gy/fraction, (Dawson et al., 2002; Pan et al., 2010).
microspheres seemed to verify this limit, under Figure 8.3b highlights the trends in Figure 8.3a
the condition of whole-liver treatments with resin at lower doses and shows the crossing of the two
spheres (Sangro et al., 2008). curves at a dose D* (~40 Gy). Of note, for a same
A subsequent step was the comparison of dose D < D*, BEDEBRT is greater than BEDRE, mean-
radioembolization with EBRT through the BED, ing that for the same effect in normal liver tissue, a
EUD, and EUBED concepts used as a guide to lower dose of EBRT would be necessary compared
define thresholds not only for whole-liver treat- with radioembolization, while the opposite holds
ments but also for lobar treatments and multicycle for D > D*.
8.4 Application of the models and examples 181

1000
120

800 100
BED EBRT
80
600 BED RE

BED (Gy)
60
BED (Gy)

400
40
BED EBRT
200
BED RE 20

0 0
0 100 200 300 400 500 0 10 20 30 40 50 60
Dose (Gy) Dose (Gy)
(a) (b)

Figure 8.3 BED as a function of absorbed dose of radioembolization and EBRT. (a) The trend in a
large interval of dose up to 500 Gy; (b) the dose interval up to 60 Gy. D* represents the dose for
which BEDRE and BEDEBRT have the same value. For a same dose lower than D*, BEDRE < BEDEBRT, while
the opposite occurs for doses higher than D*.

Table 8.2 Absorbed dose tolerance from EBRT (TD) delivered at 2 Gy/fr (Emami et al., 1991), and
correspondent BED (BEDEBRT = DEDRE) and absorbed dose values (DRE) for radioembolization

Doses or BED 1/1 whole-liver two-thirds liver One-third liver


Field (Gy) irradiation irradiation irradiation
EBRT TD5/5, EBRT 30 35 50
TD50/5, EBRT 40 45 55
EBRT = RE BED5/5,EBRT=RE 60 70 100
BED50/5,EBRT=RE 80 90 110
RE D5/5, RE 36 40 51
D50/5, RE 44 48 54

Example 8.1: Tolerance doses from EBRT extrapo-  2   2


lated to RE BEDEBRT = DEBRT  1 +  = DEBRT  1 + 
 α /β  2
In the last 25 years, several authors have presented
outcomes of liver tolerability after EBRT, includ- = 2 ⋅ DEBRT = 2 ⋅ DEBRT
ing the historical data from Emami et  al. (1991),
the analysis of Dawson et al. (2002), and the study By imposing BEDEBRT = BEDRE , the correspond-
of Pan et al. (2010) reported in the QUANTEC ing radioembolization absorbed dose is derived by
papers. The tolerance doses derived for EBRT by solving the following equation including the radio-
Emami are reported in Table 8.2 in terms of doses biology parameters previously specified (from
delivered at 2  Gy/fr and associated to 5% (TD5/5) Equations 8.7 and 8.11):
and 50% (TD50/5) probability to cause severe hepa-
titis/liver failure within 5 years. These values are
 DRE ⋅Trep 
also converted into the corresponding absorbed BEDEBRT = BEDRE = DRE  1 + 
doses in radioembolization by means of the BED  (Trep + Teff ) ⋅α / β 
concept. First, EBRT absorbed doses are converted
into BED using the following (from Equations 8.7  DRE ⋅ 2.5   D 
= DRE  1 + = DRE  1 + RE 
and 8.9):  (2.5 + 64.2) ⋅ 2   53.4 
182 The radiation biology of radioembolization

The tolerance doses of EBRT for two-thirds with the Emami values without appropriate con-
and one-third of liver irradiation could be consid- version through the BED concept because of the
ered as possible values to guide radioembolization difference in dose per fraction delivered (Emami:
treatments of the right and left lobe, respectively. 2 Gy/fr; Dawson: 1.5 Gy/fr). Similarly, the NTCP
Alternatively, if a 40 Gy absorbed dose thresh- curves corresponding to radioembolization are
old is set for whole-liver radioembolization treat- derived applying the BED conversion from EBRT
ment, the corresponding BED and absorbed dose doses, e.g., for DEBRT = 30 Gy,
in EBRT would be
 dose / fr   1.5 
 D   40  BED = DEBRT   = 30  1 +  = 52.5
BEDRE = DRE  1 + RE  = 40 ⋅  1 + = 70Gy  α /β   2 
 53.4   53.4 
 DRE ⋅Trep 
BEDRE = BEDEBRT = 70 Gy = 2 DEBRT thus, DEBRT = 35 Gy = DRE  1 + 
 (Trep + Teff ) ⋅α / β 
Example 8.2: Lyman NTCP curves derived for EBRT  DRE ⋅ 2.5 
and extrapolated for RE = DRE  1 +
 (2.5 + 64.2) ⋅ 2 
Dawson et al. (2002) used the LKB NTCP model
to interpolate clinical outcomes obtained for an  D 
EBRT scheme of 1.5 Gy, twice a day, for whole-liver = DRE  1 + RE 
 53.4 
irradiation and for partial volume irradiation as
well (Lyman, 1985; Kutcher and Burman, 1989). In thus, DRE = {[(26.7)2+53.4 ×BED]1/2}–26.7 = 33 Gy,
the case of whole-liver irradiation, the fit yielded and so on.
the following parameter values: TD50(1) = 43.3 Gy, Figure 8.4 illustrates the NTCP curves for the
m = 0.18, n = 1.1. In case of partial irradiation of Lyman model as a function of both EBRT and
two-thirds and one-third of the liver, Equation radioembolization absorbed dose, while Figure
8.33 leads to 8.5 shows the NTCP curves as a function of BED.
TD50(2/3) = 43.3·(2/3)–1.1 = 67.6 Gy, and The curves for full, two-thirds, and one-third
TD50(1/3) = 43.3·(1/3)–1.1 = 145 Gy. of irradiated liver volume can be used for irra-
It is important to note that such values, referred diation of the whole liver, the right lobe, and left
to 1.5 Gy/fr schema, cannot be directly compared lobe, respectively.

120%

100%

80%
NTCP

60%
EBRT 1/1
ER 1/1
40% EBRT 2/3
RE 2/3
20% EBRT 1/3
RE 1/3
0%
0 20 40 60 80 100 120 140 160 180 200
Dose (Gy)

Figure 8.4 NTCP curves as a function of absorbed dose for whole and partial irradiation of the
liver for EBRT and radioembolization. Uniform dose distribution within irradiated tissue is assumed.
Continuous lines represent whole liver irradiation (1/1), dashed lines represent the irradiation of 2/3
of the liver volume, and dotted lines represent the irradiation of 1/3 of the liver volume. Black curves
refer to radioembolization, gray curves refer to EBRT.
8.4 Application of the models and examples 183

120%
100%
80%

NTCP
60%
40% 1/1
2/3
20% 1/3

0%
0 50 100 150 200 250 300 350 400
BED (Gy)

Figure 8.5 NTCP curves as a function of BED assuming uniform dose distribution. Continuous line rep-
resents whole liver irradiation (1/1), dashed line represents the irradiation of 2/3 of the liver volume, and
dotted line represents the irradiation of 1/3 of the liver volume.

In the case of more selective treatments involv- Studies based on surgical resection have identified
ing a fraction ν of the whole liver, the NTCP can be the minimum remnant volume of the liver neces-
estimated either after calculating the correspond- sary to maintain function to be approximately 40%
ing TD50(ν) value (Equation 8.33) or after calculat- in patients without chronic liver disease (Kubota
ing the EUD (Equation 8.35) and using the TD50(1) et al., 1997). This figure would indicate the poten-
value. Consider, for example, the irradiation of a tial for the safe irradiation of one-third (33%) of the
segment of 100 g of normal liver tissue using radio- liver (e.g., left lobe). This represents a discrepancy
embolization, with a mean absorbed dose of 150 with the possible risk at high doses represented
Gy and with a 2100 g total liver mass: in the curves of the Lyman model extrapolated to
radioembolization (see Figures 8.4 and 8.5, curve
BED = DRE + DRE 2 /53.4 = 150 (1 + 150 /53.4 )
“1/3”). The clinical experience of radioemboliza-
= 150 ⋅ (1 + 0.0187 ⋅150 ) = 572 Gy tion seems to confirm the absence of injury when
small portions of the liver, and even the left lobe,
which corresponds to a EBRT dose (at 1.5 Gy/fr) of receive high doses. Therefore, the Lyman model
may be too conservative in the case of segmental
DEBRT = EQ1.5 = BED / (1 + 1.5 / 2) = 327 Gy
and left-lobar treatments.
and this to an EUD of (100/2100)·DEBRT = 0.048·327 A final important point to highlight regard-
= 15.6 Gy, which is associated to 0% NTCP. ing the outcomes provided by the Lyman model in
For such a small liver involvement, the corre- EBRT is that two distinct curves are obtained if a
sponding TD values are distinction is made between primary and secondary
tumors. Each curve is described by a different set of
TD50 (100 / 2100) = 43.3 × (0.048)
−1.1
= 1233Gy parameters: m = 0.12, n = 0.97, and TD50,EBRT is 40
Gy for HCC (primary tumor); m = 0.18, n = 1.1, and
TD5 (100 / 2100 ) = 30.5 ⋅ ( 0.048 )
− 1.1
= 868 Gy TD50,EBRT is 46 Gy for metastases (secondary tumor),
in the case of whole-liver irradiation. These values
that correspond to absorbed doses in RE of 314 and indicate a shift and slight deformation of the NTCP
259 Gy, respectively. curves versus higher doses in the case of metastases
This illustrates that in the extreme, when very low as compared with HCC. This is expected consider-
liver fractions are involved, the deliverable (“toler- ing that the lower liver functional reserve of the nor-
ated”) absorbed doses may reach very high values. mal liver often present in patients with HCC.
This reflects the absence of toxicity encountered in The NTCP curves generated by these parameters
selective/superselective approaches and a behavior are illustrated in Figure 8.6, with the inclusion of
similar to that of liver resection, provided that a the corresponding curves extrapolated for radio-
minimum volume of normal liver is not excised. embolization, for which TD50,RE in the case of
This is attributed to the extraordinary recovery whole-liver irradiation is 40 Gy for HCC and 44 Gy
capacity of the liver as described in Chapter 6. for metastases.
184 The radiation biology of radioembolization

120%

100%
80%

NTCP
60%
Dose EBRT met
40% Dose RE met
Dose EBRT – HCC
20% Dose RE – HCC

0%
20 30 40 50 60 70 80
Dose (Gy)

Figure 8.6 NTCP curves vs. absorbed dose of EBRT for HCC (black, continuous curve) and metastases
(black, dashed curve) and curves derived for radioembolization for HCC (gray continuous curve) and
metastases (gray dashed curve).

In summary, examples have been provided proposed, separated by a time interval sufficient to
on how to convert the EBRT parameters/doses allow liver damage recovery. In most cases, clini-
involved in the NTCP Lyman model into the cor- cians have opted for large intervals (e.g., at least 1
responding values for radioembolization. It is month) to allow the evaluation of several clinical
important to emphasize that the method shown to parameters and the regeneration of liver function-
derive radioembolization values from EBRT data ality before proceeding with additional treatment
can be nothing more than a guidance, which may cycles.
be useful in the planning of radioembolization In the case of normal liver, radiobiologi-
treatments aiming to remain on the side of safety. cal models can describe the different effects
However, the model suffers from some limita- obtained by dividing the treatment into differ-
tions due to the differences between the two irra- ent numbers of cycles at different doses per cycle;
diation modalities. For example, in some cases however, the model does not account for the time
the Lyman model predicts high toxicity if very between two consecutive cycles or the total treat-
high doses are delivered even to a small portion ment time. For tumor tissue, the total treatment
of the liver (see Figure 8.4, Example 8.2; Figures time is accounted for in the repopulation term
8.11 and 8.13), which does not agree with clinical (see Equation 8.14).
observations. Other models are able to account In general, when a treatment is composed of
for the absence of toxicity provided a minimum multiple cycles (N), each (i) giving an absorbed
portion of the liver is involved, as highlighted in dose DLi to normal liver tissue and DTi to the tumor,
Figures 8.8 and 8.9 and Example 8.4. the overall BED (BEDL for normal liver, BEDT for
For this reason, now that clinical safety data tumor) is the sum of the BED values of each cycle:
are widely available, correlation studies need to be
performed based on radioembolization dosimetry
 DLi ⋅Trep 
and clinical data, rather than extrapolation from
EBRT, allowing the derivation of self-consistent
BEDL = ∑BED = ∑ D
i
i
i
Li  1 + (T + T ) ⋅α / β 
 rep eff 
parameter values for the NTCP models.
Example 8.3: Multiple cycle approach with the
linear quadratic model
= ∑D
i
Li

 1 +
DLi 

53.4 
After the first clinical examples of hepatic toxicity
were observed following whole-liver radioem-  DTi ⋅ Trep 
bolization, less aggressive treatment methodolo-
BEDT = ∑ BED = ∑{D
i
Ti
i
Ti ⋅ 1 + 
 (Trep + Teff ) ⋅α / β 
gies were investigated. Besides lobar or selective
approaches, multiple-cycle strategies have been − K ⋅ T}
8.4 Application of the models and examples 185

In a general case of metastases (α = 0.3 Gy –1) Case 1 (Table 8.3):


and a time interval of 30 days between cycles: Radioembolization of the whole liver in two cycles
delivering 22 Gy/cycle to normal liver tissue is
BEDT = ∑ i

{DTi ⋅  1 +

Di ⋅1.5  0.693

(1.5 + 64.2) ⋅10  0.3 ⋅ 30
T} compared with a single treatment delivering the
same total dose of 44 Gy.
For the single treatment, BEDL = 44·(1 +
= ∑(D Ti ⋅ (1 + DTi ⋅ 0.0023)) − 2.3 ⋅ ( N − 1) 44/53.4) = 80 Gy, while for two cycles BEDL =
i
2 ∙ 22·(1 + 22/53.4) = 62 Gy, with a BED sparing of
where KT = 2.3 Gy represents the effect of tumor ~18 Gy (30%). At each cycle, EQ1.5 = 31.1/1.75 = 18
repopulation (i.e., the “wasted dose”). So, at least Gy, leading to a total EUD = 2·EQ1.5 = 36 Gy, asso-
with the parameters considered, the low value of ciated with an NTCP of 16%—compared with the
the wasted dose seems not to greatly influence 46 Gy EUD value of the single treatment approach,
tumor response. Alternatively, if we consider HCC with NTCP = 62%. Therefore, the use of a two cycle
assuming α = 0.01 Gy –1, the repopulation factor is therapy allows for a notable advantage in the spar-
much higher: KT = 0.693/0.01·30/30 ~69 Gy. ing of normal liver tissue.
Such modeling can be used when the treat-
ment is intentionally divided into multiple Case 2 (Table 8.4):
cycles at the planning stage, as well as in the In this case, we consider radioembolization of the
case of a retreatment, not previously foreseen left liver lobe in two cycles delivering 45 Gy/cycle
but representing a further option after a first to normal liver tissue compared with a single treat-
radioembolization therapy. However, contrary ment delivering the same total dose of 90 Gy.
to other therapies with radiopharmaceuticals The calculations follow the same process as the
that may often apply three or more cycles, the previous case, but with an additional calculation
general experience of radioembolization dem- for EUD evaluation given by EUD = ν1n (EQ1.5,1) +
onstrates the reasonable clinical feasibility of ν2n (EQ1.5,2) = 2·(0.33)1.1·47.4 = 28 Gy for two cycles,
two cycles. and EUD = νn (EQ1.5) = 0.331.1·138.2 = 41 Gy for the
Below we describe three examples (Cases 1, 2, 3) single treatment.
showing a whole liver, a left lobar, and a right lobar Also in this case, there is a notable advantage
treatment repeated in two cycles under the hypoth- for normal liver tissue, with BED and EUD sparing
eses of same administered activity and uniform of ~46% (23 Gy for BED and 13 Gy for EUD) and
dose distribution at each cycle. The questions to be a consequent NTCP reduction from high (40%) to
answered include the possible advantages of whole acceptable (3%) risk.
and lobar approaches by means of liver sparing for
a fixed dose or a possible increase of tumor irradia- Case 3 (Table 8.5):
tion for a fixed liver effect (BED). The Lyman (or This example describes radioembolization of a
LKB) model has been applied below to derive the right liver lobe in two cycles delivering a dose per
NTCP prediction. cycle to normal liver tissue that corresponds to the

Table 8.3 Comparison of the radiobiological quantities for the treatment of the whole liver in two
cycles as compared with one single cycle with a same total absorbed dose of 44 Gy (case 1).

Case 1: Whole liver First cycle Second cycle Single treatment


Volume % 100% 100% 100%
Dose RE (Gy) 22 22 44
BEDi (Gy) 31 31 80.3
EQ1.5,i = EUDi (Gy) 18 18 46
EUD tot (Gy) 36 (Δ = –29%) 46
BED tot (Gy) 62 (Δ = –29%) 80
NTCP % 16% 63%
186 The radiation biology of radioembolization

Table 8.4 Radiobiological quantities for the treatment of the left liver lobe in two cycles as compared
with one single cycle with a same total absorbed dose of 90 Gy (case 2).

Case 2: Left lobe 1st cycle 2nd cycle Single treatment


Volume (%) 33 33 33
Dose RE (Gy) 45 45 90
BEDi (Gy) 83 83 242
EQ1.5,i (Gy) 47 47 138
EUDi (Gy) 14 14 41
EUD tot (Gy) 28 (Δ = –46%) 41
BED tot (Gy) 50 (Δ = –46%) 72
NTCP (%) 3% 40%

Table 8.5 Comparison of the radiobiological quantities for the treatment of the right liver lobe in two
cycles as compared with one single cycle with a same total absorbed dose of 48 Gy (case 3).

Case 3: right lobe 1st cycle 2nd cycle Single treatment


Volume (%) 67 67 67
Dose RE,i (Gy) 30 30 48
BEDi (Gy) 46 46 91
Dtot RE (Gy) 59 (+23%) 48
Liver

EQ1.5,i (Gy) 26 26 52
EUDi (Gy) 17 17 34
EUDtot (Gy) 33.5 33.5
NtCP % 10% 10%
Dose RE,i (Gy) 92 92 150
tumor

BEDi (Gy) 112 112 201


Dtot RE (Gy) 185 (Δ = 23%)
BEDtot (Gy) 221 (Δ = 10%) 201

same BED obtained for a single treatment with an 150·0.0023) = 201 Gy for the single treatment, and
absorbed dose of 48 Gy. BEDT, tot = 2·92.3·(1 + 92.3·0.0023) – 2.3 = 221 Gy
A single dose to the liver lobe of 48 Gy corre- in two cycles.
sponds to a BED = 48·(1+48/53.4) = 91 Gy, which The BED gain is, therefore, 20 Gy correspond-
when split in two cycles corresponds to BED = 46 ing to a gain of 10% in BED. This is lower than the
Gy/cycle and a dose of 30 Gy/cycle. For the same increase in the total dose, which is 23%.
effect (risk) to the liver, the cumulative dose to the Figure 8.7 illustrates the possible cumulative
liver lobe is 59 Gy as compared with 48 Gy of a sin- dose increase as a function of the absorbed dose
gle treatment, indicating a possible increase of the delivered to the liver by radioembolization when
cumulative activity by a factor of 1.23, and conse- splitting the treatment in two or three cycles.
quently a same dose increase to the tumor. Although three cycles may be clinically impracti-
To quantify the effect on tumor, consider a cal, it is interesting to see the trend and to have a
delivery of a dose DT = 150 Gy to the tumor for rough idea of the benefit realized by this technique.
a single treatment approach. If the dose increase
per cycle previously derived is applied, a dose per Example 8.4: Various models compared
cycle of DT,1cycle = 92 Gy/cycle is obtained. Setting The previous examples described the extrapola-
an interval of 30 days between the two cycles, tion of useful information from the EBRT experi-
the repopulation term is TK = 2.3 Gy and the ence (Lyman model) obtaining a first estimate of
correspondent BED values are BEDT = 150·(1 + the response to radioembolization. As previously
8.4 Application of the models and examples 187

60%

50%

40%

30%

A% 20%
2 cycles
10%
3 cycles

0%
0 20 40 60 80 100
Dose RE in 1 cycle (Gy)

Figure 8.7 Possible cumulative dose increase as a function of the absorbed dose delivered to the liver
by radioembolization when splitting the treatment in 2 or 3 cycles.

evidenced, this procedure may lead, in some cases, fr 50


to improbable predictions. Other models may be a= = 7.48
σfr 2
more suited to describe radioembolization out-
comes from clinical data. f −  fr 50
In the ensuing discussion, the LKB, the par- b=
σfr  √ 2
allel architecture, and the Walrand models are
considered comparing their predictions in exem- with the fit parameters: fr50 = 0.497, σfr =  0.047,
plificative cases and highlighting the volume d50 = 43 Gy (absorbed dose in RE), k = 3. The
effect that each incorporates. Of special interest last two parameters were obtained by imposing
is the irradiation of small portions of the liver that, for same BED = DEBRT·Erel,EBRT =  DRE·Erel,RE,
and the different effects related to the use of resin p(DEBRT) = p(DRE).
as compared with glass spheres characterized by Walrand model
a higher tolerability and lower responsiveness.
NTCP = 1/(1+TD50/D)γ
Assuming for simplicity a uniform dose distri-
where
bution D in a volume fraction ν of the normal liver
tissue, the parameters needed to apply for the dif- TD50 = (25.2 + 22.1·(1–exp(–2.74·msA)))/(ν–0.4)0.584
ferent models consist of the following: γ = 13.7·(ν)2 + 30.6·ν –8.41
Lyman model:
D is the mean dose (regardless of uniformity) and
EUD = νn × D msA is the microsphere-specific activity (0.05 kBq for
resin microspheres, 2.5 kBq for glass microspheres).
 EUD − TD50 
NTCP = 1 2 − 1 2 erf  

Figure 8.8 illustrates the NTCP curves of the
 mTD50 2  parallel architecture model as a function of dose D
for the case of uniform irradiation of different liver
with n = 1.1, m= 0.18, TD50 = 43 Gy, erf = error portions. Similarly, Figure 8.9a and 8.9b shows the
x
2 same NTCP curves for the Walrand model, for
function =
π ∫e dt
t2
msA of 0.05 kBq (resin spheres, Figure 8.9a) and
0
2.5 kBq (glass spheres, Figure 8.9b). Despite some
Parallel architecture model: differences, the volume effect is quite evident in
both cases, and for irradiation of 40% (or less) of
NTCP = ½·[erf(a) + erf(b)] if f > 0.497 or
the total volume both models predict a null NTCP
NTCP = ½·[erf(a) – erf(b)] if f < 0.497
irrespective of the absorbed dose. Thus, cases with
where f = ν/(1 + d 50/D)k is the fraction of organ less than 40% of liver involvement will not be
damaged and presented in this section because only the Lyman
188 The radiation biology of radioembolization

120% 100%
67%
100%
50%
80% 45%
40%

NTCP %
60%

40%

20%

0%
0 50 100 150 200 250 300
Dose RE (Gy)

Figure 8.8 NTCP curves for the parallel model in the case of uniform dose to the entire volume of
normal liver tissue (black curve), 67% of normal liver (gray curve), 50% of normal liver (dot-dashed
curve), 45% of the normal liver (dotted curve), and 40% of the normal liver (dashed curve).

120% 120%

100% 100%

80% 80%
NTCP

60% 60% 100% – 2.5


100% – 0.05
NTCP

67% – 0.05 67% – 2.5


40% 50% – 0.05 40% 50% – 2.5
45% – 0.05 45% – 2.5
20% 40% – 0.05 20% 40% – 2.5

0% 0%
0 50 100 150 200 250 300 0 50 100 150 200 250 300 350 400 450 500
Dose RE (Gy) Dose RE (Gy)
(a) (b)

Figure 8.9 NTCP curves for the Walrand model as a function of mean dose, irrespective of uniformity,
delivered to the whole liver (black continuous curve), 67% of normal liver (gray curve), half of the liver
(dotted curve), 45% of the normal liver (dashed curve), and 40.1% of the liver (dot-dashed curve).
(a) curves for msA= 0.05 kBq (resin spheres) and (b) those for 2.5 kBq (glass spheres).

model still shows curves predicting the possible kBq are quite similar in the case of 67% of liver
toxicity if a high dose is delivered. In particular, an involvement, while the model of Walrand for
example already discussed is the radioemboliza- 2.5 kBq shows a higher tolerance as compared
tion of the left lobe for which Lyman still indicates with resin spheres, in agreement with clinical
a possible complication risk (see Figures 8.4 and observations. On the contrary, at lower volume
8.5 for one-third of the liver volume). fractions, such as for irradiation of 50% of nor-
mal liver, the predictions of the different models
8.4.1 NTCP PREDICTIONS UNDER diverge.
THE HYPOTHESIS OF Table 8.6 summarizes the parameters, the
UNIFORM IRRADIATION absorbed doses, and the evaluations of risk
derived for several representative cases using
Figure 8.10 directly compares the NTCP curves the three NTCP models, under the hypothesis of
of the different models for a uniform dose deliv- uniform dose. In particular, Cases 1A–D repre-
ered to 67% (Figure 8.10a) and to 50% (Figure sent whole-liver treatments delivering different
8.10b) of normal liver tissue. Interestingly, the doses to the parenchyma: 27 Gy, where all mod-
models of Lyman, Parallel, and Walrand for 0.05 els predict no toxicity; 40 Gy, which is considered
8.4 Application of the models and examples / 8.4.1 NTCP predictions under the hypothesis 189

120% 120%
100% 100%
80% 80%
60%
NTCP

NTCP
60%
40% P v67%
40% P v50%
W 0.05 – v67%
20% W 2.5 – v67% W 0.05 – v50%
20% W 2.5 v50%
L v67%
0% L v67%
0%
0 50 100 150 200 250 300 0 50 100 150 200 250 300
Dose RE (Gy) Dose RE (Gy)
(a) (b)

Figure 8.10 NTCP curves of the different models under the hypothesis of a uniform dose delivered
to 67% (a) and to 50% (b) of normal liver tissue. The black curve represents the parallel model and the
gray curve represents the Lyman model. The dotted and dashed curves are the model by Walrand for
0.05 kBq and 2.5 kBq microspheres, respectively.

Table 8.6 Parameters, absorbed doses (uniform), and evaluations of risk derived for several
representative cases using the different NTCP models
Irradiated liver portion Whole liver right lobe Left lobe Segment

Cases 1A 1B 1C 1D 2A 2B 2C 3A 3B 4B
Legends in the figure WL27 WL40 WL58 WL80 RL 37 RL 50 RL105 LL37 LL80 S250
V% of NTL involved 100% 100% 100% 100% 67% 67% 67% 33% 33% 10%
NTL dose (Gy) 27 40 58 80 37 50 105 37 80 250
LKB model EUD (RT) 23 40 69 114 23 35 153 11 40 61
NTCP 0% 34% 100% 100% 0% 16% 100% 0% 33% 99%
Parallel Damaged V% 20% 45% 71% 87% 26% 41% 64% 13% 30% 9%
model NTCP 0% 14% 100% 100% 0% 2.8% 100% 0% 0% 0%
Walrand TD50 38 38 38 38 61 61 61 n.e. n.e. n.e.
model msA NTCP 5% 62% 97% 100% 5% 24% 96% 0% 0% 0%
0.05 kBq
Walrand TD50 63.7 64 64 64 102 102 102 n.e. n.e. n.e.
model msA NTCP 0% 2% 31% 87% 0% 1% 54% 0% 0% 100%
2.5 kBq
Abbreviations: WL27, WL40, WL58, WL80, whole liver receiving a mean dose of 27, 40, 58, 80 Gy, respectively; RL37,
RL50, RL105, right liver lobe receiving a mean dose of 37, 50, 105 Gy, respectively; LL37, LL80, left liver lobe receiving
a mean dose of 37, 80 Gy, respectively; S250, liver segment receiving a mean dose of 250Gy; V%, liver volume%, mSA
(0.05 kBq for resin, 2.5 kBq for glass microspheres); n.e. is not evaluable; NTL, normal liver.

a threshold for possible toxicity by some authors 3B refer to treatment of the left lobe delivering
(Cremonesi et al., 2008); 58 Gy, which is asso- a low dose of 37 Gy, with no risks foreseen by
ciated to different toxicity probabilities by the any model, and a dose of 80 Gy, where only the
models; and 80 Gy, which has been suggested Lyman model shows nonnegligible risk. Finally,
as a limit for safety by the empiric approach in Case 4 reports the predicted toxicity for a highly
the operators’ manual of the resin spheres. Cases irradiated segment (250 Gy).
2A–C consider the irradiation of the right lobe Details are presented in Table 8.6, while Figure
at doses of 37 Gy, indicating safety, 50 Gy, asso- 8.11 illustrates a visual comparison of these results.
ciated with acceptable risk, and 105 Gy, with For whole-liver treatment, all the models pre-
high risk foreseen by all models. Cases 3A and dict a maximum risk at 80 Gy. Maximum risk
190 The radiation biology of radioembolization

100%

90%

80% LKB

70% Parallel

60% Walrand 0.05


NTCP %

50% Walrand 2.5

40%

30%

20%

10%

0%
WL 80 WL 58 WL 40 WL 27 RL 105 RL 48 RL 37 LL 80 LL 37 S250

Figure 8.11 NTCP predictions by the Lyman model (dark gray bar), the Parallel model (light gray bar),
the Walrand model for msA of 0.05 (white bar) and for msA of 2.5 (black bar), according to the cases
included in Table 8.6 (WL80, WL58, WL40, WL27 refer to whole liver with uniform dose of 80, 58, 40,
and 27 Gy, respectively; RL105, RL48, RL37 refer to right lobe irradiation with uniform dose of 105, 48,
and 37 Gy, respectively; LL80, LL37 refer to left lobe irradiation with uniform dose of 80, and 37 Gy,
respectively; S250 refers to a segment irradiation with uniform dose of 250 Gy).

is also seen at 58 Gy, except in the case of glass 8.4.2 NTCP PREDICTIONS FOR
microspheres due to their decreased toxicity. CLINICAL CASES WITH
Of note, at 40 Gy and even 27 Gy the Walrand NONUNIFORM IRRADIATION
model for resin microspheres indicates higher
risk compared with the other models. The When considering real clinical situations, the dose
Walrand model for resin spheres is in fact more distribution—as evidenced by posttreatment
conservative than even the Lyman model for imaging—is always nonuniform. To include this
large liver volumes irradiated at low doses. This nonuniformity in the models, the DVH {νi, Di}
relationship can also be observed for right lobe needs to be derived and used to calculate EUD for
radioembolization up to ~50 Gy, as illustrated the Lyman model:
in Figure 8.10a. Also for 50% liver irradiation
and low doses, this model predicts nonnegligi- EUD = ( Σi νi ⋅ (Di )1/n )n s
ble NTCP values higher than the other models
up to ~65 Gy. However, for higher doses more and the fraction of damaged organ for the parallel
agreement is seen. For example, in the case of architecture model:
right lobe radioembolization and a dose of
105  Gy, all models indicate unacceptable risks, f = Σi νi / (1 + 43 / Di )3
being lower in the case of glass microspheres,
but higher than would normally be clinically Oddly, the Walrand model requires only the
acceptable (54%). Finally, as already pointed out, evaluation of the mean absorbed dose to the vol-
for left lobe radioembolization and segmental ume treated, without taking into account the mac-
approaches, only the Lyman model indicates the roscopic absorbed-dose uniformity.
presence of risk, with NTCP values as high as Below, the DVH of four patients has been used
100%. However, as already outlined, the Lyman to compare the predictions of the models. Patients
model is not reliable for left lobe or segmental 1 and 2 underwent right lobe radioemboliza-
radioembolization. tion with similar mean doses to the treated lobe
8.4 Application of the models and examples / 8.4.2 NTCP predictions for clinical cases 191

(48  and 44 Gy, respectively), but different dose nonuniformity observed by posttreatment imag-
distributions and thus, different NTCP. Patient  3 ing (patient 1: RL Dm48; patient 3: RL Dm105;
underwent right lobe radioembolization with patient 4: LL Dm80) to NTCP predictions neglect-
a mean dose of 105 Gy, and patient 4 underwent ing nonuniformity, i.e., accepting the hypothesis of
a left lobe treatment with a mean dose of 80 Gy. a uniform dose equal to the mean absorbed dose
Figure 8.12a and 8.12b shows the DVHs of patients (patient 1: RL,D48; patient 3: RL,D105; patient 4:
1 and 4, respectively. Table 8.7 gathers the results LL,D80).
of the evaluations using each model with the mean As previously evidenced, the models by Walrand
dose specified in place of uniform dose. Figure predict the same values with or without uniformity.
8.13 gives an immediate comparison of the NTCP Using the other models, the histogram shows the
predictions. typical effect of nonuniformity. In the case of the
Figure 8.14 compares the NTCP predic- right lobe with a mean dose of 105 Gy, the parallel
tions in patients 1, 3, and 4 accounting for the architecture model changes from suggesting 100%

120 120
Pt4, LL, Dm80
Pt1, RL, Dm48 100
100
80 V (%) 80
60
V (%)

60
40 40

20 20

0 0
0 100 200 300 400 0 100 200 300 400 500 600
Dose (Gy) Dose (Gy)
(a) (b)

Figure 8.12 DVHs of patient 1 (a), who received a mean dose of 48 Gy to the right lobe (Pt1, RL, 48
Gy), and patient 4 (b), who received a mean dose of 80 Gy to the left lobe (Pt4, LL, 80 Gy).

Table 8.7 NTCP evaluations from different models from the DVHs of patients
Irradiated liver portion right lobe Left lobe
Patients and legends (Figure 8.13) Pt1 Pt2 Pt3 Pt4
rL, Dm48 rL, Dm44 rL, Dm105 LL, Dm80
NtL V% involved 67 67 67 67
NtL mean dose rE (Gy) 48 44 105 80
LKB model EQ1.5 (EBRT) 70 55 252 86
EUD (EBRT) 60 48 216 68
NtCP 98% 73% 100% 100%
Parallel model Damaged V% 22% 22% 36% 16%
NtCP 0% 0% 0% 0%
Walrand model TD50 61 61 61 n.e.
(0.05 kBq) NtCP 20% 20% 96% 0%
Walrand model TD50 102 102 102 n.e.
(2.5 kBq) NtCP 1% 1% 54% 0%
Abbreviations: Pt1, RL, Dm48 = patient 1, right liver lobe irradiation with a mean dose of 48 Gy; Pt2, RL, Dm44 =
patient 2, right liver lobe irradiation with a mean dose of 44 Gy; Pt3, 2/3, Dm105 = patient 3, right liver lobe irradia-
tion with a mean dose of 105 Gy; Pt4, 1/3, Dm48 = patient 4, left liver lobe irradiation with a mean dose of 80 Gy;
EQ1.5 is the corresponding EBRT dose released at 1.5 Gy/fr; V%, the liver volume%; mSA (0.05 kBq for resin, 2.5 kBq
for glass microspheres); NTL, the normal liver; n.e., not evaluable.
192 The radiation biology of radioembolization

100%

90% LKB
Parallel
80%
Walrand 0.05
70%
Walrand 2.5
60%
NTCP %

50%

40%

30%

20%

10%

0%
RL, Dm48 RL, Dm44 RL, Dm105 LL, Dm80

Figure 8.13 Comparison of the NTCP predictions for patient 1 (RL, Dm48: right lobe, mean dose 48
Gy), patient 2 (RL, Dm44: right lobe, mean dose 44 Gy), patient 3 (RL, Dm105: right lobe, mean dose
105 Gy), and patient 4 (LL, Dm80: left lobe, mean dose 80 Gy). The dark gray bars are associated to
the Lyman model, the light gray bars to the Parallel model, the white bars to the Walrand model for
msA = 0.05 kBq, and the black bars to the Walrand model for msA = 2.5 kBq.

100%
90%

80%
70% LKB
Parallel
60%
Walrand 0.05
NTCP %

50%
Walrand 2.5
40%

30%
20%
10%

0%
RL, D105 RL, Dm105 RL, D48 RL, Dm48 LL, D80 LL, Dm80

Figure 8.14 NTCP predictions by different models accounting for non-uniformity or accepting the
hypothesis of a uniform dose equal to the mean absorbed dose. Data shown for patient 1 (RL, D48: right
lobe, uniform dose of 48 Gy; RL,Dm48: right lobe, same mean dose of 48 Gy but accounting for non-
uniformity), patient 3 (RL,D105: right lobe, uniform dose of 105 Gy; RL,Dm105: right lobe, same mean
dose of 105 Gy but accounting for non-uniformity), and patient 4 (LL,D80: left lobe, mean dose 80 Gy;
LL,Dm80: left lobe, same mean dose of 80 Gy but accounting for non-uniformity). The dark gray bars
are associated to the Lyman (LKB) model, the light gray bars to the parallel model, the white bars to the
Walrand model for msA = 0.05 kBq, and the black bars to the Walrand model for msA = 2.5 kBq.

NTCP to a feasible therapy when nonuniformity the Lyman model: for left lobe radioembolization
is taken into account. However, the Lyman model with a mean dose of 80 Gy, the EUD for uniform
maintains a 100% NTCP because of its conservative dose is 34 Gy, while accounting for nonuniformity
characteristics, especially at high doses. The other the EUD reaches ~68 Gy. This leads to the prediction
two cases, however, show an unexpected effect for of feasibility in the case of uniform dose distribution
8.5 Summary for clinical applications 193

5000

4000

EQ 1.5 and BED (Gy)


EQ1,5
BED
3000

2000

1000

0
0 100 200 300 400 500
Dose RE (Gy)

Figure 8.15 BED and EQ1.5 curves derived as a function of dose of radioembolization. The continuous
line represents BED, and the dashed line represents EQ1.5, i.e., the dose of EBRT released at of 1.5
Gy/fraction.

and high risk with nonuniform distribution. This is the clinical practice of radioembolization. While
the opposite of what was observed with the parallel medical physicists and scientists will be able to apply
architecture model for the right lobe in both the 105 aforementioned radiobiologic models to clinical
and 48 Gy cases. The predictions of the Lyman model therapy, the broader concepts presented below will
are also contrary to clinical observations, which sug- be appreciated by other members of the radioem-
gest that while hot spots of nonuniformity raise the bolization treatment team, including interventional
mean dose, they should lower the global injury and radiologists and nuclear medicine physicians.
decrease NTCP.
Figure 8.15 should help to explain this point by 1. Liver hypertrophy and resectability: Lessons
showing the BED and EQ1.5 (the correspondent from surgery show that liver tissue is able to
EBRT dose released at 1.5 Gy/fr) as a function of regenerate if a certain portion of the liver is
radioembolization dose. The calculation of the embolized (i.e., excluded from blood circula-
EUD includes the conversion of the radioemboli- tion) or even excised. This property of the liver
zation absorbed dose to each voxel into the corre- is used to increase the efficacy of liver resec-
sponding EBRT dose (via the BED). However, the tion by pretreatment portal-vein embolization,
DVH of real patients can show high doses following which artificially augments the future remnant
radioembolization (e.g., up to ~300 Gy for patient 1, liver volume. The final result is a lower occur-
RL, Dm48; up to ~500 Gy for patient 4, LL, Dm80), rence of hepatic complications/insufficiency
that correspond to extremely high doses of EBRT. after partial hepatectomy.
For example, 200 Gy in radioembolization should There are many studies from surgery regarding
correspond to 1000 Gy in EBRT, and 500 Gy fol- liver resectability, in particular the minimum
lowing radioembolization corresponds to 5000 Gy portion of liver remnant necessary to avoid
of EBRT. The validity of the Lyman model, which hepatic damage or failure. In summary, the
is a phenomenological derivation, can only be sup- literature reports that, in a major hepatectomy,
ported at much lower ranges of doses of EBRT and a remnant: (1) ≤20–25% could be at high risk of
with nonuniformities that are not extreme. complication in patients with normal liver func-
tion (Chun et al., 2008; Lin et al., 2014; Truant et
al., 2015; Vauthey et al., 2000); (2) ≥40% should
8.5 SUMMarY FOr CLINICaL guarantee safety in patients without chronic liver
aPPLICatIONS disease (Kubota et al., 1997); (3) ≤ 40% and 55%
would represent considerable risk for failure in
The aim of this section is to assemble the main patients heavily treated by chemotherapy, and
radiobiological issues described in the previous sec- patients with cirrhotic liver, respectively (Narita
tions and provide direct insight into their effect on et al., 2012; Lin, 2014). These guidelines can be
194 The radiation biology of radioembolization

aptly applied for partial liver radioembolization 4. Safety of lobar and selective radioemboliza-
(see point 4 below). tion: According to current models, for seg-
2. Hypertrophy and radioembolization: The mental or selective radioembolization for the
presence of hypertrophy emerges also in the treatment of tumors with minimal involve-
follow-up imaging of many patients who have ment, NTCP is negligible at reasonable clini-
underwent radioembolization as an intrin- cal absorbed doses. Such models adhere to
sic defense against the radiation damage to clinical observations in both radioemboliza-
liver cells. Such a property increases the liver tion and surgery (see point 1 of this section).
function, offering a major resource that can be In particular, the minimum recommended
fully exploited when partial liver irradiation or remnants for a safe liver resection in patients
multiple-cycle strategies are applied. with different disease status/pathologies offer
The regeneration capability of the liver is not guidelines for partial liver radioembolization.
included in any of the mathematical models In fact, the worst consequence induced by
discussed in this chapter but should appear in irradiation is cell killing, which can be associ-
phenomenological observations, with curves ated with tissue removal, i.e., surgery/resec-
describing higher tolerability than expected. tion. Therefore, the data summarized in point
So, in the parallel and the Walrand theoretical 1 above can be related to safe partial-liver
models, it might be necessary to add a further irradiation with radioembolization, which
element that takes into account this phenom- could involve 60% of the liver (i.e., sparing
enon. Alternatively, as more toxicity data 40% of normal liver tissue) for patients with
becomes available, some parameters might normal liver function. In patients heavily
be adjusted to account for a radiosensitivity treated with chemotherapy and in cirrhotic
lower than predicted. In other words, slightly patients, 60% (i.e., 40% spared) and 45% (i.e.,
less conservative models might better reflect 55% spared) thresholds can be appropriately
observed results, with a shift toward higher applied from surgical resection data. Even
doses for liver damage. adding a margin of prudence, the barrier
3. Multiple-cycle approach and time interval of no risk imposed by the parallel and the
between cycles: A multicycle radioemboliza- Walrand models at volume fractions lower
tion strategy is certainly of help to reduce the than 40%, irrespective of the dose, is nicely
risk of toxicity or to increase the dose delivered coherent with the experience of surgery. This
to the tumor. The gain in terms of absorbed refers to patients with normal liver function
dose or BED has been shown in Example 8.4 or minimal previous chemotherapy treatment,
for the linear quadratic model. while for patients with liver disease (HCC,
In initial multicycle therapy trials, the time substantial previous chemotherapy) a further
interval between cycles was an open question. margin for safety may be taken into account.
Some authors proposed just a few days between 5. Sparing effect of nonuniform dose:
cycles, which could be a retreatment of a same Nonuniformity weakens the effect of radia-
target volume or a separately treatment of right tion. This comes from a common logic and
and left lobes. This short time could be accept- from the EBRT data (Kassis and Adelstein,
able from a radiobiological perspective, as the 2005). Since the worst insult resulting
repair of radiation damage occurs in a time from radiation is cell death, as radiation
frame on the order of hours (see Section 8.2.1). dose increases beyond the threshold neces-
However, the liver can do more than repair sary for cell killing, there is no biological
injured cells; it is capable of regeneration, and impact of further increasing the radiation
so short intervals have been replaced by at least dose. Therefore, in the case of nonunifor-
30- to 40-day intervals. Longer intervals are mity, there may be tissue areas with wasted
needed to induce a countervailing hypertro- energy delivered (due to an absorbed dose
phy, with the intent to recover as much of the well above the threshold for cell killing), and
liver functionality as possible between treat- other areas where the dose is insufficient to
ment cycles. provoke the same damage as the mean dose.
References 195

The formalism of the Lyman and the parallel


models include the possibility of accounting 8.6 CONCLUSIONS
for nonuniformity. In fact, the NTCP values
associated with uniform and nonuniform In this chapter, the basic principles of radiation biol-
doses for a same mean dose do differ by these ogy have been illustrated, together with the most
models. However, it must be emphasized that important models that could be used to describe
the parallel model correctly provides lower the outcomes of radioembolization. Some models
NTCP values in the case of nonuniformity, are derived from the experience of EBRT and can
while the Lyman model can fail in certain be very useful so long as limitations in their appli-
cases, predicting higher NTCP values for uni- cability are considered. Other models have been
form irradiation when very high doses exist in developed in the context of radioembolization, and
the dose map (see the discussion of the results can be more suitable to describe clinically observed
for pt1 in Figure 8.14 [RL,D48 vs. RL,Dm48] effects.
and Figure 8.15). In general, many concepts have been illus-
6. Conservativeness and risks to be added: trated to give the reader useful instruments and
While conservatism is often taken in treat- confidence with the different models and formal-
ment planning for radioembolization of isms available in the literature. Once assimilated,
patients with HCC, there are factors that sug- all these concepts should be applied to clinical
gest a precautionary attitude for all patients radioembolization data with a critical and con-
may be warranted. Most patients receiving scientious attitude. With increasing dosimetric
radioembolization are not naïve to therapy data and clinical evidence, it will be possible to
but have already received at least two lines build more robust models allowing better pre-
of hepatotoxic chemotherapy. With this in dictivity and personalization of radioemboliza-
mind, the differences between NTCP curves tion treatments, following the example of EBRT.
related to HCC and to metastatic patients In this sense, continued collection of toxicity and
(Figure 8.6) could be less than predicted. This efficacy data should be encouraged, as well as the
consideration relates also to what has been performance of personalized pre- and posttreat-
reported in point 1 above. Thus, a distinction ment 3D dosimetry with the highest possible level
between two NTCP curves lower than that of accuracy.
provided by Dawson and Pan (Dawson et al.,
2002; Pan et al., 2010) for EBRT should not be All models are wrong, but some are useful (George
unexpected. E. P. Box)
7. Toxicity evaluation: The collection of toxic-
ity data represents a milestone for correla- REFERENCES
tion analysis with doses and radiobiological
quantities. To further refine radiobiologic Antipas, V., Dale, R.G., Coles, I.P. (2001). A
models for radioembolization moving theoretical investigation into the role of
forward, reliable methods with appropri- tumour radiosensitivity, clonogen repopu-
ate timing, completeness, and uniformity lation, tumour shrinkage and radionuclide
among researchers is needed. In particular, RBE in permanent brachytherapy implants of
the analysis of the cholinesterase is strongly 125I and 103Pd. Phys Med Biol 46:2557–2569.
recommended as a method to evaluate liver Baechler, S. et al. (2008). Extension of the biolog-
function damage, for patient screening and ical effective dose to the MIRD schema and
follow-up (Meng et al. 2013). This is a most possible implications in radionuclide therapy
reliable indicator for liver injury, commonly dosimetry. Med Phys 35(3):1123–1134.
used in surgical and hepatological disci- Brenner, D.J. et al. (1998). The linear-quadratic
plines, although less common in nuclear model and most other common radiobio-
medicine and interventional radiology. The logical models result in similar predictions
evaluation of indocyanine or xenobionts is of time-dose relationships. Radiat Res
also good alternative methods. 150:83–91.
196 The radiation biology of radioembolization

Burman, C. et al. (1991). Fitting of normal tissue treated with 90Y-loaded glass micro-
tolerance data to an analytic function. Int J spheres: preliminary results. J Nucl Med
Radiat Oncol Biol Phys 21:123–135. 53(2):255–263.
Chiesa, C. et al. (2015). Radioembolization of Garin, E. et al. (2015). Personalized dosimetry
hepatocarcinoma with 90Y glass micro- with intensification using 90Y-loaded glass
spheres: development of an individualized microsphere radioembolization induces pro-
treatment planning strategy based on dosim- longed overall survival in hepatocellular car-
etry and radiobiology. Eur J Nucl Med Mol cinoma patients with portal vein thrombosis.
Imaging 42(11):1718–1738. J Nucl Med 56(3):339–346.
Chun, Y.S. et al. (2008). Comparison of two meth- Garin, E. et al. (2016). Clinical impact of (99m)
ods of future liver remnant volume measure- Tc-MAA SPECT/CT-based dosimetry in the
ment. J Gastrointest Surg 12(1):123–128. radioembolization of liver malignancies with
Cremonesi, M. et al. (2008). Radioembolisation (90)Y-loaded microspheres. Eur J Nucl Med
with 90Y-microspheres: dosimetric and radiobi- Mol Imaging 43(3):559–575.
ological investigation for multi-cycle treatment. Jackson, A. et al. (1995). Analysis of clinical com-
Eur J Nucl Med Mol Imaging 35(11):2088–2096. plication data for radiation hepatitis using a
Cremonesi, M. et al. (2014). Radioembolization of parallel architecture model. Int J Radiat Oncol
hepatic lesions from a radiobiology and dosi- Biol Phys 31:883–891.
metric perspective. Front Oncol 4:210. Jackson, A., Kutcher, G.J., Yorke, E.D. (1993).
Dale, R.G. (1996). Dose-rate effects in targeted Probability of radiation-induced complica-
radiotherapy Phys Med Biol 41:1871–1884. tions for normal tissues with parallel architec-
Dale, R.G., Jones, B., Sinclair, J.A. (2000). Dose- ture subject to non-uniform irradiation. Med
equivalents of tumour repopulation during Phys 20:613–625.
radiotherapy: the potential for confusion Jones, L.C., Hoban, P.W. (2000). Treatment plan
Br J Radiol 73:892–894. comparison using equivalent uniform biologi-
Dawson, L.A., Lawrence, T.S., Ten Haken, R.K. cally effective dose (EUBED). Phys Med Biol
(2001). Partial liver irradiation. Semin Radiat 45(1):159–170.
Oncol 11:240–246. Kassis, A.I., Adelstein, S.J. (2005). Radiobiologic
Dawson, L.A. et al. (2002). Analysis of radia- principles in radionuclide therapy. J Nucl Med
tion-induced liver disease using the Lyman 46(Suppl 1):4S–12S.
NTCP model. Int J Radiat Oncol Biol Phys Kong, M., Hong, S.E. (2015). Optimal follow-up
53(4):810–821. duration for evaluating objective response
Dawson, L.A., Ten Haken, R.K. (2005). Partial vol- to radiotherapy in patients with hepatocel-
ume tolerance of the liver to radiation. Semin lular carcinoma: a retrospective study. Chin J
Radiat Oncol 15(4):279–283. Cancer 34(2):79–85.
Emami, B. et al. (1991). Tolerance of normal tissue Kubota, K. et al. (1997). Measurement of liver vol-
to therapeutic irradiation Int J Radiat Oncol ume and hepatic functional reserve as a guide
Biol Phys 21:109–122. to decision-making in resectional surgery for
Flamen, P. et al. (2008). Multimodality imag- hepatic tumors. Hepatology 26(5):1176–1181.
ing can predict the metabolic response of Kutcher G., Burman, C. (1989). Calculation
unresectable colorectal liver metastases to of complication probability factors for
radioembolization therapy with Yttrium-90 non-uniform normal tissue irradiation: the
labeled resin microspheres. Phys Med Biol effective volume method Int J Radiat Oncol
53(22):6591–6603. Erratum in: Phys Med Biol Biol Phys 16(6):1623–1630.
2014;59(10):2549–2551. Lin, X.J., Yang, J., Chen, X.B., Zhang, M., Xu,
Garin, E. et al. (2012) Dosimetry based on M.Q. (2014). The critical value of remnant liver
99mTc-macroaggregated albumin SPECT/ volume-to-body weight ratio to estimate pos-
CT accurately predicts tumor response and thepatectomy liver failure in cirrhotic patients.
survival in hepatocellular carcinoma patients J Surg Res188(2):489–495.
References 197

Lyman, J.T. (1985). Complication probability as Strigari, L. et al. (2011). Dosimetry in nuclear
assessed from dose-volume histograms. medicine therapy: radiobiology applica-
Radiat Res 104:S13–S19. tion and results. Q J Nucl Med Mol Imaging
Manda, G. et al. (2015). The redox biology net- 55(2):205–221.
work in cancer pathophysiology and thera- Truant, S. et al. (2015). Liver function following
peutics. Redox Biol 5:347–357. extended hepatectomy can be accurately
Meng, F. et al. (2013). Assessment of the value of predicted using remnant liver volume to body
serum cholinesterase as a liver function test for weight ratio. World J Surg 39(5):1193–1201.
cirrhotic patients. Biomed Rep 1(2):265–268. Vauthey, J.N. et al. (2000) Standardized mea-
Millar, W.T. (1991). Application of the linear-quadratic surement of the future liver remnant prior to
model with incomplete repair to radionuclide extended liver resection: methodology and
directed therapy. Br J Radiol 64:242–251. clinical associations. Surgery 127(5):512–519.
Mitsuishi, Y., Motohashi, H., Yamamoto, M. Walrand, S., Hesse, M., Jamar, F., Lhommel, R.
(2012). The Keap1–Nrf2 system in cancers: (2014a). A hepatic dose-toxicity model
stress response and anabolic metabolism. opening the way toward individualized
Front Oncol 2. Available from: http://dx. doi. radioembolization planning. J Nucl Med
org/10.3389/fonc.2012. 55(8):1317–1322.
Narita, M. et al. (2012). What is a safe future liver Walrand, S. et al. (2014b). The low hepatic toxic-
remnant size in patients undergoing major ity per Gray of 90Y glass microspheres is
hepatectomy for colorectal liver metastases linked to their transport in the arterial tree
and treated by intensive preoperative chemo- favoring a nonuniform trapping as observed
therapy? Ann Surg Oncol. 19(8):2526–2538. in post-therapy PET imaging. J Nucl Med
Niemierko, A., Goitein, M. (1993). Modeling 255:135–140.
of normal tissue response to radiation: the Wigg, D.A. (2001). Applied Radiobiology and
critical volume model. Int J Radiat Oncol Biol Bioeffect Planning. Madison, WI: Medical
Phys. 25:135–145. Physics Publishing.
O’Donoghue. (1999). Implications of nonuniform Withers, H.R., Taylor, J.M.G., Maciejewski, B.
tumor doses for radioimmunotherapy: equiva- (1988). Treatment volume and tissue toler-
lent uniform dose. J Nucl Med 40:1337–1341. ance. Int J Radiat Oncol Biol Phys. 15:751-759.
Pajonk, F., Vlashi, E., McBride, W.H. (2010). Withers, H.R. (1992). Biological basis of radiation
Radiation resistance of cancer stem cells: therapy for cancer. Lancet 339(8786):156–159.
the 4 Rs of radiobiology revisited. Stem Cells Yorke, E.D. et al. (1992). Probability of radiation-
28(4):639–648. induced complications in normal tissues with
Pan, C.C. et al. (2010). Radiation-associated parallel architecture under conditions of
liver injury. Int J Radiat Oncol Biol Phys 76(3 uniform whole or partial organ irradiation.
Suppl):S94–S100. Radiother Oncol 26:226–237.
Sangro, B. et al. (2008). Liver disease induced by Yorke, E.D. et al. (1999). Can current models
radioembolization of liver tumors: descrip- explain the lack of liver complications in
tion and possible risk factors. Cancer Y-90 microsphere therapy? Clin Cancer Res
112(7):1538–1546. 5:3024s–3030s.
Strigari, L. et al. (2010). Efficacy and toxicity Yorke, E.D. et al. (2001). Modeling the effects of
related to treatment of hepatocellular car- inhomogeneous dose distributions in normal
cinoma with 90Y-SIR spheres: radiobiologic tissues. Semin Radiat Oncol 11(3):197–209.
considerations. J Nucl Med 51(9):1377–1385.
9
Microsphere deposition, dosimetry,
radiobiology at the cell-scale, and
predicted hepatic toxicity

STEPHAN WALRAND

9.1 Introduction 199 9.8 Microspheres biodistribution studies


9.2 Scales in liver radioembolization 200 in liver 206
9.3 Introduction to Monte Carlo 9.9 Hepatic arterial tree modeling 207
methods 201 9.10 Microsphere transport modeling 208
9.4 Dose deposition around a β source 203 9.11 Microsphere distribution simulation 209
9.5 Voxel-based absorbed dose 204 9.12 Microsphere distribution and hepatic
9.6 Intralobule dosimetry from Monte toxicity 211
Carlo simulations in translation 9.13 Application of the hepatic toxicity
invariant trapping 204 model 212
9.7 Intralobule dosimetry from Russell’s 9.14 Conclusions 215
law in translation invariant trapping 205 References 215

electron Monte Carlo (MC) tracking. Because


9.1 INtrODUCtION MC electron transport is computationally bur-
densome, Gulec et al. (2010) assumed that all the
Over the past decade, it has become well estab- hepatic lobules shared the same microsphere trap-
lished that hepatic toxicity per Gy is significantly ping pattern enabling the use of a fast reflective
different between 50 Bq/sphere resin and 2500 boundary technique. In this translation invariant
Bq/sphere glass 90Y microspheres (Kennedy et al., setup, the simulation did not clearly establish a dif-
2007). An overview of the similarities and differ- ference in hepatic toxicity per Gy between the two
ences between resin and glass microspheres is pre- microsphere devices.
sented in Chapter 1. The hepatic toxicity per unit However, experimental microscopy studies of
absorbed dose of glass microspheres is about one- microsphere distribution have revealed strongly
third of that observed in external beam radiother- nonuniform microsphere trapping (Pillai et al.,
apy (EBRT) (Dawson et al., 2001). 1991; Roberson et al., 1992; Campbell et al., 2000;
Gulec et al. (2010) performed the first simula- Kennedy et al., 2004). Chiesa et al. (2011) suggested
tion of cell-scale dosimetry applied to compare the that the lower hepatic toxicity per Gy observed
effects of hepatic radioembolization using resin with glass microspheres could be due to a more
and glass microspheres. Gulec et al. (2010) used nonuniform microsphere distribution owing to

199
200 Microsphere deposition, dosimetry, radiobiology at the cell-scale

their lower number, resulting in sparing of more length and ≈1.2 mm diameter (Figure 9.1). A por-
regions of normal hepatic parenchyma. tal triad, consisting of a bile duct, a portal venule,
This chapter reviews the recent experimental and a few arteries (2.4 on average; Crawford et al.,
and theoretical developments that have provided a 1998), is located at each corner of the prism. The
better understanding of this difference. Some pre- six portal triads are each shared by three lobules,
dictions of the hepatic toxicity in liver radioembo- resulting in a total number of triads ≈2 × 106. The
lization as a function of microsphere number and hepatic arterial tree consists of approximately 21
target liver volume fraction are also provided. vessel bifurcations or about 221 ≈ 2 × 106 terminal
The author would like to emphasize that if these arterioles. Compared with all other tissues, the
developments prove the hepatic toxicity per unit hepatic lobules have the unique feature to be fed by
absorbed dose decreases with decreasing micro- both arterial and venous sources. After injection
spheres number, then the tumor response per unit via a hepatic artery branch, the microspheres that
dose is also theoretically expected to decrease. This are larger than the intralobule arteriole diameter
fact is confirmed with clinical observation. As a are predominantly trapped in linear clusters in the
result, the theoretical study of the impact of micro- triad arteries. Thus, the most uniform activity dis-
sphere number on therapy efficacy requires mod- tribution already exhibits a heterogeneity periodic
eling of the microsphere distribution in tumor, pattern of ≈1.5 mm scale corresponding to the
which is a challenging problem due to the anarchic length of each lobule.
nature of tumor vasculature. Blood outflow in normal hepatic structure is
ensured by a single vein located at the center of the
lobule—the central vein. As the primary venous
9.2 SCaLES IN LIVEr drain, integrity of the central vein is essential to
raDIOEMBOLIZatION preserve the blood flow and thus lobule viability.
On the other hand, as the nutrient and oxygen dif-
The human adult liver is a lattice of ≈106 indepen- fusion range in soft tissue is approximately 500 μm,
dent functional subunits called lobules (Gulec et al., one or two preserved portal triads are likely enough
2010). Each lobule is a hexagonal prism of ≈1.5 mm to keep the lobule alive, although this has not yet

Central vein

Hepatic sinusoids

Branch of the
hepatic artery

Branch of the
portal vein

Bile duct
and ductule

Single surviving portal triad

Figure 9.1 Schematic representation of lobule. (Courtesy of Will McAbee, Educational Resource
Center, College of Veterinary Medicine at University of Georgia, Athens, GA.) Because hexagon
side-to-side distance is about 1200 μm, almost all lobule structures are closer than 500 μm from small
intralobule arterioles or venules (highlighted) still transporting blood from only one surviving triad.
9.3 Introduction to monte carlo methods 201

been validated in an experimental in vivo model. explains why living donor liver transplantation
The absorbed dose in the vicinity of a microsphere (LDLT) can safely survive when only 33% of the
reaches several hundred Gy; therefore, a portal triad liver remains while 90% of the cells in the residual
trapping one or more microspheres will suffer from liver undergo proliferation or mitosis (Haga et al.,
local microscale radiation necrosis. 2008). This also explains why liver is one of the
The dose delivered by a 90Y loaded micro- most radioresistant tissues.
sphere quickly decreases with the distance, by Is it safe to kill two-thirds of the liver volume by
a factor ≈1000 from the microsphere boundary irradiation? Obviously not! Partial liver irradiation
up to 0.5  mm. This might suggest that the cen- in EBRT teaches us that killing 60% and 40% of the
tral vein or portal triad empty of microspheres is liver volume by irradiation gives a normal tissue
quite preserved from lethal irradiation due to dis- complication probability (NTCP) of 99% and 50%,
tance alone. However, the maximal range of the respectively (Dawson et al., 2001). The major dif-
90Y β-particle is about 11 mm, and consequently, ference with surgical resection is that immediately
all the lobule structures are also irradiated by after irradiation the surviving liver volume has
the microspheres trapped in the 600 closest sur- no free space to regenerate, has to handle toxins
rounding lobules. The following sections will show released by dying cells, and has to recycle necrotic
that the mean absorbed dose in a lobule free of a tissue while maintaining homeostasis.
microsphere, but surrounded by lobules contain-
ing a constant number of microspheres, is only
20% lower than if that lobule contained the same 9.3 INtrODUCtION tO MONtE
number of microspheres. As a result, microsphere
CarLO MEtHODS
trapping heterogeneities on the centimeter scale,
rather than the millimeter scale, are thus required
to preserve lobule structures from lethal radiation. MC methods often appear quite obscure to the non-
Using typical radiation dosages and number physicist. MC methods are based upon repeated,
of infused microspheres for both resin and glass numerous random drawings (or sampling) accord-
microspheres (Chapter 1), the average number of ing to a specific probability distribution in order
spheres per triad is 16 resin and 1 glass micro- to numerically solve a mathematical or a physical
sphere, delivering about 40 and 120 Gy to the liver problem. Let us illustrate this concept with a sim-
parenchyma, respectively. As a result of the ran- ple example.
dom transport of the microspheres through the Typically, to assess the value of π, one would
arterial tree by the flow of blood, microsphere clus- start from the relation π / 4 = tan−1 (1) to develop
ter sizes are distributed around the mean number the inverse tangent function in the Taylor series
of microspheres per triad. In comparison, a typi- and to numerically compute the terms of the series.
cal 250 MBq 18F-fludeoxyglucose positron emis- More sophisticated series expansions of π have
sion tomography (18FDG-PET) scan corresponds been developed allowing fast computation of tril-
to 105 18FDG molecules per lobule (assuming 4.5% lions of decimal digits. Besides this computational
of uptake in the liver; Mettler and Guiberteau, method, there are two simple experimental meth-
2012). In contrast to radioembolization, this high ods to estimate π.
number of FDG molecules per lobule dramatically The first one, often performed in elemen-
smooth transport fluctuations. This explains why tary school, is to surround a disc by a rope and
the FDG distribution in liver appears, and is, much to compute the ratio between the length of the
more uniform than that of microspheres. rope and the diameter of the disk. A drawback of
In contrast to other tissues, liver regeneration is this method is that it requires an accurate length
not dependent on a small group of stem cells, but measurement.
is carried out by proliferation of its intact mature A second method which was one of the first
cells (Michalopoulos and DeFrances, 1997). applications of MC is (1) draw equidistant and
Hepatocytes can proliferate almost without limit, parallel lines on a floor by moving a pen along the
but more remarkably they have the capacity to side of a rectangular rule, the opposite side being
proliferate while simultaneously performing all successively shifted to the last drawn line, (2) set
essential functions needed for homeostasis. This a wood stick along the small side of the rule and
202 Microsphere deposition, dosimetry, radiobiology at the cell-scale

cut it off at the same width. Using this setup, GL integer division by m. This generator has a period
Leclerc Comte de Buffon mathematically proved p, which is lower than m, i.e., after p generations the
that if the stick were randomly dropped, the prob- sequence of p numbers is repeated. Although the
ability that the stick will lie across a line is 2 / π random number stream produced using Equation
(Schroeder, 1974). By dropping the stick numer- 9.1 is deterministic, a set of generated numbers less
ous times and counting how many times it crossed than its period cannot be distinguished from a true
a line, it is thus possible to get an estimation of π random set using simple statistical tests.
(Figure 9.2). The elegance of this experimental MC Nowadays, fast uniform pseudorandom number
estimation of π is that it involves no measurement generators are based on Mersenne twister theory
of length. (Saito et al., 2008, http://www.agner.org/random/).
The modern success of MC methods originated They are fast, have an extremely long period (up
from the development of uniform pseudorandom to 211213–1), succeed in all known statistical tests,
number generators, i.e., algorithms for generat- and are thus well adapted to MC methods. Specific
ing a sequence of numbers that cannot be distin- probability distribution can be obtained from the
guished from a true random sequence, such as that uniform pseudorandom generator using the trans-
obtained using a roulette wheel. Historic random formation of variable or rejection methods (Press
generators were based on the recurrence equation et al., 2007).
(Press et al., 2007): MC methods have the capacity to increase the
speed of some numerical computations and to
ni+1 = (a  ni +  c ) %m (9.1)
allow simulation of physical process governed
where a, c, m are positive integers and % represents by probabilistic laws. Various codes are used in
the modulus operation, i.e., the remainder of the nuclear medicine (Ljungberg et al., 2013). In this

Figure 9.2 Schematic representation of Buffon’s needle experiment showing the lying positions of a
stick randomly dropped 20 times on the ground. When the length of the stick matches the distance
in between the lines, the fraction of positions crossing a line tends to 2/π when the dropping number
increases.
9.4 Dose deposition around a β source 203

chapter, MC methods are used for three purposes: approximation using the simple equation (Russell
(1) computation of the absorbed dose delivered et al., 1988):
around a β point source, (2) growing of a syn-
thetic arterial tree, and (3) microsphere transport  r 1
DR (r ) =  D0  1 −    2 (9.2)
through the arterial tree.  R r
The dose D(r) quickly decreases with the
distance  r. However, when the activity is spread in
a large region of tissue, it is better to consider the
9.4 DOSE DEPOSItION arOUND function 4 πr 2   D(r ). This function describes the
a β SOUrCE dose received in a point from all the activity located
at a distance r when the uptake is uniform. In this
Since electron trajectories are governed by the case, one can clearly see that the mean absorbed
probabilistic laws of quantum electrodynamics dose coming from the activity localized within the
(QED), the only way to compute the dose deliv- 0.7 mm radius surrounding region (dark gray area in
ered from a β source is to simulate numerous elec- Figure 9.4b) is much smaller than that coming from
tron trajectories, electron by electron, and to sum the farther activity (light gray area in Figure 9.4b).
the energy spatially deposited by each trajectory. The small differences between the different MC
Nearly all MC codes simulate electron trajectories computations mainly arise from the different set-
in discrete small steps (Figure 9.3). ups: Cross et al. (1992) modeled a 90Y point source in
Figure 9.4a shows the dose D(r) delivered in a water, Gulec et al. (2010) modeled a 32-μm-diameter
medium as a function of the distance r to a small spherical activity distribution inside a soft tissue
90Y source computed using different MC codes, equivalent medium, and Paxton et al. (2012) also
except the dashed line that represents an analytical modeled the actual microsphere density.

New e– emission

Direction θ φ?
energy E?

New e– direction θ φ?
Q
ED

Free step (θ, φ)


D = D + ΔE Yes

Q Q Q
ED ED ED
No Yes No

Interaction? Which? New e– energy > ε?

Figure 9.3 Symbolic representation of MC simulation of electron histories. After the random drawing of
an emission direction, the e – freely travels a small step in this direction, then the occurrence of an inter-
action with the medium and eventually the new properties of the e – are randomly drawn according to
the cross sections computed from quantum electrodynamics (QED) theory. The energy deposited by the
interaction is summed to the absorbed dose, and when the residual e – energy becomes negligible (≤ε)
the tracking of the electron is stopped and a new e – emission is initiated. For the sake of clarity, emis-
sions of secondary particles (such as photons produced by bremsstrahlung, recoil electrons, electron–
positron pair creations, etc.) were not represented. Dices are loaded according to the QED rules.
204 Microsphere deposition, dosimetry, radiobiology at the cell-scale

1.E+03 6.E–03
Cross et al. 1992 (MC ETRAN code)
1.E+02 Russell’s law 1988
Gulec et al. 2010 (MCNPX code) 5.E–03
1.E+01 Paxton et al. 2012 (MC EGSNRC code)

4πr2 D [ηGy/h × cm2]


1.E+00 4.E–03
D [ηGy/h × cm2]

1.E–01
3.E–03
1.E–02

1.E–03 2.E–03

1.E–04
1.E–03
1.E–05

1.E–06 0.E+00
0.01 0.10 1.00 10.00 0 1 2 3 4 5 6 7 8 9 10
r [mm] r [mm]

(a) (b)

Figure 9.4 (a) Dose deposition around a small 50 Bq 90Y source in soft medium. (b) Dose delivered in
r = 0 coming from spherical shell of radius r in a uniform 90Y distribution. Cross et al. (1992): ETRAN
simulation for point source in water. Gulec et al. (2010): MCNPX simulation for a 30 μm spherical
source in soft tissue equivalent medium. Paxton et al. (2012): EGSNRC simulation for glass 25 μm
microsphere surrounded by liver tissue.

Equation 9.4 is not analytically computable and


9.5 VOXEL-BaSED aBSOrBED one could be tempted to numerically compute it as
DOSE
N −1 N −1 N −1 N −1 N −1 N −1

Assuming a liver of uniform density, a fast and easy


K u ,v ,w =
1
  ∑∑∑   ∑∑∑ 
N 6 m= 0 n=0 s =0 i =0 j =0 k =0

method to compute the voxel-based absorbed dose (9.5)



(uH + (m − i ) h) + (vH + (n − j )h) + (wH + ( s − k )h) 
2 2 2
from a known voxel based activity is to convolve it DR 
 
by a voxel dose kernel K. The mean absorbed dose
Dijk in a voxel (i,j,k) is simply where H is the voxel size and h = H/N.
However, much more efficiently, Equation 9.5
Dijk =   ∑ K i −m , j −n , k − s    N mns (9.3)
can be reduced into a triple summation:
N N N
1
∑∑∑ (N − i ) N − j (N − k )  ( )
mns
K u ,v ,w =  
N 6 i =− N j =− N k =− N
where Nmns is the number of decays occurring in (9.6)
the voxel (m,n,s). The kernel element K u , v , w rep-
(uH + ih ) + (vH + jh ) + (wH + kh )
2 2 2
resents the mean absorbed dose delivered into the DR (  )
voxel (u,v,w) when one decay occurred in a position Other methods of determining voxel dose ker-
averaged in the whole voxel (0,0,0). nels are presented in Chapter 12.
The voxel dose kernel K can be computed by
MC or more easily by integrating Russell’s law
(Equation 9.2): 9.6 INtraLOBULE DOSIMEtrY
FrOM MONtE CarLO
1 r 1 SIMULatIONS IN
K u ,v ,w =
V
 
∫∫∫ r
y ∈Vuvw
dy  
V ∫∫∫ r
x ∈Vooo
(9.4)
traNSLatION INVarIaNt
r r r
traPPING
(
dx DR y − x )
Gulec et al. (2010) computed the intralobule dose
where Vuvw is the voxel (u,v,w) domain while V is distribution by MC for a translation invariant 90Y
the volume of the voxels. loaded microsphere distribution in which all the
9.7 Intralobule dosimetry from Russell’s law 205

lobules of the liver trap microspheres with the microspheres was roughly rescaled to get a mean
same pattern. Two microsphere distributions were liver absorbed dose of 40 and 120 Gy for the 50 Bq
modeled: 24 × 50 Bq microspheres in each portal and 2500 Bq microsphere models, respectively
triad and 1 × 2500 Bq microsphere in every other (Kennedy et al., 2004; Lau et al., 2012). For the
portal triad, both distributions corresponding to a triad structures, the absorbed doses are given for
mean liver absorbed dose of ≈64 Gy. a triad containing, or not, a 2500 Bq microsphere.
To boost the computation speed, the reflective The results of these simulations show that
boundary method was used: only one lobule is in both models, all the portal triads, including
modeled in the MC simulation and when a tracked those not containing any microspheres, receive
electron reached the lobule boundary, it underwent a lethal mean absorbed dose which is in discrep-
a reflection back into the lobule in order to account ancy with the low toxicity observed in clinical
for the cross-fire effect of the surrounding lob- therapy.
ules. Similarly to Figure 9.4b, the MC simulations
showed that four-fifths of the absorbed dose to the
central vein and about one-half of the absorbed 9.7 INtraLOBULE DOSIMEtrY
dose to the triad vein came from the microspheres FrOM rUSSELL’S LaW IN
trapped in the surrounding lobules.
traNSLatION INVarIaNt
Table 9.1 shows the mean absorbed dose to the
lobule structures derived from the Gulec et al.
traPPING
(2010) MC simulations. In order to better cor-
respond to the clinical practice, the number of Even to compute the absorbed dose distribu-
tion by MC electron tracking in a single lobule,
Table 9.1 Mean absorbed dose in the different Gulec et al. (2010) boosted the computation speed
hepatic structures by using the reflective boundary method that
D for 50 D for 2500 required identical microsphere trapping in all
Bq/ms, 15 Bq/ms, 2 ms lobules. Computing the absorbed dose distribu-
ms per triad every other tion by tracking the electrons inside all the 106
tissue (Gy) triad (Gy) lobules, each surrounded by lobules having dif-
ferent trapping patterns, is far beyond the capac-
Liver 40 120
ity of the state-of-the-art computing technologies.
Hepatocytes 39 120
A much faster alternative is to use the Russell
Central vein 37 109
dose distribution. Table 9.2 shows the compari-
Triad bile duct 70 109–320 son of the intralobule dosimetry obtained by
Triad vein 68 109–501 Gulec et al. (2010) by MC electron tracking and
Triad artery 118 109–636 that obtained using the Russell dose distribution

Table 9.2 Comparison of mean absorbed doses computed with MC and from the Russell law

1 × 2500 Bq ms in every other triad


24 × 50 Bq ms in each triad artery artery
russell
MC (Gulec et al., (Walrand et al., MC (Gulec et al., russell (Walrand
tissue 2010) (Gy) 2014a) (Gy) 2010) (Gy) et al., 2014a) (Gy)
Liver 64 63 64 65
Hepatocytes 63 63 64 65
Central vein 59 58 58 60
Triad bile duct 112 118 58–171 60–187
Triad vein 109 113 58–167 60–182
Triad arteriole 188 206 58–339 60–377
206 Microsphere deposition, dosimetry, radiobiology at the cell-scale

6 78 168 402 582 856 1148 6 78 168 402 582 856 1148
480 160

420 140 Artery


Artery
360 Bile 120 Bile
Vein Vein
300 1 × 2500Bq-ms/triad 100 15 × 50Bq-ms/triad
Hepa Hepa
D [Gy]

D [Gy]
240 80 Central
Central
180 40 Gy 60 40 Gy

120 40

60 20

0 0
>0 >1.3 >2.6 >3.9 >5.2 >6.5 >7.8 >8.1 >0 >1.3 >2.6 >3.9 >5.2 >6.5 >7.8 >8.1
r [mm] r [mm]
(a) (b)

Figure 9.5 Absorbed dose to the different lobule structures coming from the microspheres trapped
in triad arteries located farther than i × 1.3 mm to the lobule center. (a) 1 × 2500 Bq microsphere
in each triad artery. (b) 15 × 50 Bq microspheres in each triad artery. The number of portal triads
included between the two concentric shell of radius i × 1.3 mm and (i + 1) × 1.3 mm is indicated on
the upper axis.

(Equation 9.2). The relative deviation is about


4% for hepatocytes and central vein and about 9.8 MICrOSPHErES
10% for structures containing or in contact with BIODIStrIBUtION StUDIES
microspheres. IN LIVEr
Figure 9.5 shows the computed absorbed doses
to the different lobule structures coming from Early studies (Pillai et al., 1991; Roberson et al.,
the microspheres trapped in the triad arteries 1992) on microspheres biodistribution were per-
located at a distance farther than i x 1.3 mm. Two formed in explanted rabbit livers after radio-
scenarios were simulated: 1 × 2500 Bq micro- embolization with 27-μm-diameter polystyrene
sphere per triad artery corresponding to a mean microspheres. At a mean number of four micro-
liver absorbed dose of 130 Gy, typical of a clini- spheres per triad, i.e., in between clinical resin and
cal therapy with glass microspheres (Figure 9.5a), glass microspheres radioembolization, Pillai et al.
and 15 × 50 Bq microspheres per triad artery (1991) found some clusters larger than 25 micro-
corresponding to a mean liver absorbed dose of spheres. Some reports followed on cluster gather-
40 Gy, typical of a clinical therapy using resin ings studied in two-dimensional (2D) sections of
microsphere (Figure 9.5b). explanted human liver tumors (Campbell et al.,
This simulation shows that, for a surrounding 2000; Kennedy et al., 2004).
translation invariant microspheres trapping, a Recently, Högberg et al. (2014, 2015a, 2015b)
8-mm- and a 3-mm-diameter sphere free of micro- conducted a first real three-dimensional (3D)
spheres are required to keep the absorbed dose scanning of the microsphere clusters in 16 biop-
inside the central lobule below 40 Gy for the 120 sies of a normal human liver tissue explanted 9
and 40 Gy scenarios, respectively. days after radioembolization using resin micro-
The fact that the needed heterogeneity scale is spheres. The autoradiography of the explanted
larger in the setup in which fewer microspheres tissue (Figure 9.6a) showed an extremely nonuni-
are injected is compatible with statistical fluctua- form microsphere distribution with 1-cm-scale
tions resulting in the transport dynamics of the subregions of very low or of high microsphere
microspheres from the catheter tip up to the termi- densities. The 16 biopsies displayed 125 single
nal triad arteries. Prediction of these fluctuations microspheres and 277 clusters containing a total of
requires a model of the hepatic arterial tree. 3736 microspheres. Two different types of clusters
9.9 Hepatic arterial tree modeling 207

(b)

5 cm
(a) (c)

Figure 9.6 (a) Autoradiography of normal liver tissue explanted 9 days after resin microspheres radio-
embolization. (b and c) Slice of 2 of the 275 clusters found in the 16 biopsies performed in the liver
tissue. (b) Linear cluster of microsphere sequentially trapped in terminal triad artery. (c) Central slice
of a globular cluster with microspheres gathered inside a larger artery. (Courtesy of Dr. Högberg and
of Dr. Bernhardt.)

were identified: linear clusters (Figure 9.6b), corre- and thus a small imaging voxel. Using a 5 μm
sponding to microspheres sequentially trapped in a voxel size, the reconstruction of the whole liver
( )
3
terminal triad artery, and globular clusters (Figure will have to be performed using a 6 × 104 matrix,
9.6c), corresponding to microspheres gathered in which is still far beyond the limits of conventional
larger arteries, likely at branching nodes where the computers. In addition, automatic analysis pro-
artery splits into two smaller arteries. The mean grams still exhibit segmentation issues that have to
cluster size (or equivalently the mean number of be manually addressed when two vessel branches
microspheres per triad) in the biopsies was 9.2. The touch. This alone could represent a monumental
largest cluster had a globular shape and contained task when one considers the 4 × 106 vessel branches
453 microspheres. Large globular clusters were to be segmented. However, Debbaut et al. (2012,
found in artery generations 13–19, with a maximal 2014) obtained a very impressive vascular tree seg-
frequency in the 17th and 18th generations. mentation up to the sinusoid level but on a limited
2 mm × 2 mm × 2 mm sample size.
Significant improvements were obtained this
last decade in the mathematical modeling of the
9.9 HEPatIC artErIaL trEE hepatic vascular tree. Three different approaches
MODELING are competing: constrained constructive optimi-
zation (CCO), deterministic geometric construc-
The spatial resolution of current human in vivo tion, and angiogenesis-based construction (the
computed tomography (CT) is a little less than reader can find an exhaustive literature survey and
0.5 mm. Although this spatial resolution continu- a discussion of these three approaches in Schwen
ously improves, in vivo imaging of the whole hepatic and Preusser, 2012).
arterial tree down to 40 μm (diameter of triad Currently, CCO, introduced by Schreiner and
arteries) will likely remain inaccessible, especially Buxbaum (1993), is a very promising approach
considering the inability to completely eliminate (Schwen and Preusser, 2012). Briefly, CCO is an
motion effects. Ex vivo corrosion casting is a pow- MC process where the arterial tree is updated by
erful technique that, in theory, should be able to randomly drawing in the liver a free node that
achieve this goal, especially when combined with a is afterward connected to the closest branch of
high-dose industrial CT capable of submicrometer the arterial tree (see the video demo at http://
spatial resolution (Cnuddea and Boone, 2013). w w w.mevis-research.de/~oschwen/research/
However, detailed simulation of the micro- talks/20120823-BerlinISMP-iCCO.pdf). The ini-
sphere transport dynamics requires an accurate tial tree consists of a major hepatic vessel network
assessment of the vessels’ diameter and curvature, obtained from CT arteriography. The optimization
208 Microsphere deposition, dosimetry, radiobiology at the cell-scale

step consists of designing new branch bifurcations The simulations showed that the microsphere
to minimize the total vascular volume taking into partition at an arterial node does not follow that
account that the vessel radii are, at each iteration of the blood flow. In addition, it depends on the
step, constrained to ensure an equal blood flow to microsphere position in the vessel lumen prior
all the lobules. to the node, the flow acceleration phase, and the
Recently, Schwen and Preuser (2012) built a bifurcation angles of the daughter vessels. These
realistic arterial tree, but only supplied 10,000 simulations were confirmed in an experimental
nodes. Assuming the viscosity was independent model (Richards et al., 2012, 2013).
to the vessel radius, which is only valid for radius After having crossed several bifurcations, one
larger than 150 μm, the workload for generating can expect that the particles are more or less evenly
N nodes is of the order O(N2 ln(N)). Thus, the gen- distributed in the vessel lumen. Microsphere injec-
eration of the whole-liver arterial tree will require tion is often performed slowly during several
60,000-fold more computation time. Taking into cardiac cycles, the impact of which is therefore
account the radius dependence of the viscosity averaged. In a steady state, Kennedy et al. (2010)
will still significantly increase the workload. showed that for a uniform inflow, the local parti-
tions between the four daughter vessels (1, 2, 3, 4
in Figure 9.7) were (0.26, 0.20, 0.29, 0.25) and (0.14,
0.32, 0.36, 0.18) for the blood flow and resin micro-
9.10 MICrOSPHErE traNSPOrt spheres, respectively. Thus, local microsphere par-
MODELING titioning in the nodes of daughter vessels (1,2) and
(3,4) was (0.30, 0.70) and (0.67, 0.33), respectively.
These microsphere partitions must be corrected
Kennedy et al. (2010) and Basciano (2010) mod- for small blood flow differences. At the first order,
eled fluid dynamics and microsphere transport i.e., for the assumption that the microspheres fol-
in the four major branches of a hepatic arte- low the blood flow, the correction is
rial tree derived from the population-represen-
tative morphological data. The computations
were performed under the hypothesis of dilute Pi ms =
pims
 ms
 pi +  
1 − pims( )      (9.7)
microsphere suspension, i.e., the presence of pibf  pibf
 1 − pibf( ) 
microspheres does not impact the fluid dynamics
ms bf
and the interaction between microspheres can be where pi and pi are the simulated local parti-
neglected. Simulations were performed not only tion of daughter i for the microspheres and for the
ms
in steady flow, but also in transient dynamics blood flow, respectively. Pi is the corrected parti-
by introducing in the equations a hepatic pres- tion, i.e., rescaled to equal daughter blood flow.
sure waveform also derived from population- After correction using Equation 9.7, microsphere
representative data. partitions become (0.25, 0.75) and (0.63, 0.37). Note

35 40.0
PB = PD
Parabolic inlet
Local particle exit percentage (%)

Branch vessel 30 35.0


PB = 1.2PD Uniform inlet
Local flow percentage (%)

30.0
25
Particle
injection 25.0
20
plane 20.0
Parent vessel 15
15.0
4 10
Inflow 10.0
3 5 5.0
Daughter vessels 0 0.0
1 2
Daughter 1 Daughter 2 Daughter 3 Daughter 4 Daughter 1 Daughter 2 Daughter 3 Daughter 4

Figure 9.7 (Left) Arterial branches modeled. Middle: percentage of incoming blood flow exiting indi-
vidual daughter vessels. (Right) Percentage of incoming particles exiting individual daughter vessels.
(Reprinted from Int J Radiat Oncol Biol Phys, 76, Kennedy et al., Computer modeling of yttrium-90-
microsphere transport in the hepatic arterial tree to improve clinical outcomes, 631–637, Copyright
(2010), with permission from Elsevier.) Note that for the uniform inlet, even when the blood flow is
lower, the particles preferably go into the two most curved bifurcation, i.e., daughters 2 and 3.
9.11 Microsphere distribution simulation 209

that for the two nodes (1,2) and (3,4), the micro- Under this constraint, the node position and the
sphere partition is always greater in the bifurcating folding of the existing vessel that minimizes the
vessel. total length of the vessels were selected. Minimizing
Basciano (2010) reported a computing time of the total vessel length rather than the total vessel
about 60 hours per microsphere tracked through volume avoided the recomputation of all the vessel
the three nodes of the model using a quad core radii that is needed after each new lobule connec-
CPU. Simulating millions of microspheres through tion in order to ensure an equal blood flow to all the
the 20 successive nodes of a liver will remain chal- lobules, saving considerable computational time.
lenging for many years. When the arterial tree is built, the blood flow
of all vessel branches was computed to ensure an
equal blood flow to all the terminal triad arteries.
The probability of each terminal triad artery trap-
9.11 MICrOSPHErE
ping a microsphere was computed by following, in
DIStrIBUtION SIMULatION reverse, the artery path from the triad to the cath-
eter tip. At each node, the probability was mul-
In order to achieve a reasonable computation time, tiplied by the local microsphere partition of the
Walrand et al. (2014a) built a full 3D hepatic arterial considered bifurcation, rescaled by its local blood
tree using a simplified CCO scheme, i.e., the total ves- flow partition using Equation 9.7.
sel length was optimized rather than the total vessel Triad arteries were randomly populated under
volume. Microsphere dynamics and transport were the dilute microsphere suspension assumption,
modeled by a simple random selection at each node i.e., microsphere by microsphere according to the
of the daughter vessel crossed by the microsphere. probability associated with a given triad. After each
The main trunk, composed of the eight artery microsphere delivery, the trapping probability of
branches feeding the eight liver segments, was the triad was reduced on order to account for the
manually drawn according to the standard liver reduction of blood flow by partial embolization.
morphology. The 2 × 106 triad arteries were succes- As lobule triads have on average 2.4 arteries, each
sively randomly selected in the liver volume and the 1300 μm in length, the reduction was designed
closest existing vessel was identified (Figure 9.8). such that the trapping probability linearly vanishes
The position of the connection node in this vessel after 300 microspheres.
was constrained to be closer to the trunk than to Figure 9.9 shows a slice comparison of simu-
the selected triad. This constraint avoids retrograde lated 2500 Bq microsphere distributions (Figure
artery vessels that are not physiologically present. 9.9b and c) delivering 120 Gy to the liver versus a

1
3

Figure 9.8 One iteration step of the simplified CCO arterial tree generation. 1. Random selection of a
free lobule. 2. Identification of the closest existing vessel branch. 3. Determination of the new connec-
tion node position and of the existing branch folding which minimizes the total vessel length.
210 Microsphere deposition, dosimetry, radiobiology at the cell-scale

Virtual arterial tree Simulated 90Y distribution

50% – 50%

60% – 40%

(a) (b)

Simulated 90Y distribution Patient 90Y TOF-PET Phantom 90Y TOF-PET

(c) (d) (e)

Figure 9.9 (a) 3D rendering of virtual arterial tree after generation of the first 1500 vessels. (b and
c) Glass microsphere distribution with a 120 Gy average liver dose from a virtual arterial tree using
50%–50% (b) and 60–40% (c) microsphere relative-partition probability between two daughter ves-
sels. Both slices were convolved with a blurring kernel to match PET spatial resolution. (d) Typical 90Y
TOF PET slice in normal liver of a patient treated with glass microspheres at a 120 Gy average left liver
dose. Note the similar granularity of glass microsphere distribution shown in (c) and (d). (e) TOF PET
imaging of hot sphere phantom with the same acquisition time and same 90Y-specific activity as shown
in patient image in (d). (Reprinted in black and white from Walrand et al., J Nucl Med, 5, 135–140,
2014a.)

typical time of flight (TOF) 90Y PET acquisition of a terminal triad arteries, and globular clusters,
patient (Figure 9.9d) and of a hot spheres phantom shown in Figure 9.6c, are not present in the simu-
(Figure 9.9e). The patient was treated with glass lation. The largest cluster contained 158 micro-
microspheres with a 120 Gy average dose to the spheres, which is threefold less than that observed
left liver lobe, while the phantom was filled with by Högberg (2015b).
an identical specific background activity. More In order to also simulate globular clusters,
information on 90Y PET imaging can be found in Högberg (2015b) developed an arterial tree includ-
Chapter 11. ing an exponentially decreasing diameter of
Figure 9.10 shows the cumulated cluster size arterial branches from the main trunk up to the
distribution observed by Högberg (2015b) from terminal triad arteries as observed by Debbaut
biopsies of normal liver tissue explanted 9 days et al. (2012, 2014). Three variable parameters were
after radioembolization with resin microspheres optimized to obtain concordance between simu-
(see Figure 9.6a). The best agreement with the lated and in vivo microsphere distributions: (1) a
model of Walrand et al. (2014a) was obtained for combined artery coefficient of variation (ACV)
an asymmetric microsphere partition probability parameter for the inner diameter of all arterial
of 64%–36% at the bifurcation nodes in line with generations throughout the virtual tree structure
the dynamic transport simulations (Basciano, that controls the microsphere flow distribution at
2010; Kennedy et al., 2010). Although the cluster the nodes, (2) the hepatic tree distribution volume
size distribution is well predicted in this model, (HDV) parameter, and (3) the embolization (EMB)
all of the microsphere clusters are located in the parameter that reduces the arterial diameter.
9.12 Microsphere distribution and hepatic toxicity 211

1.0
0.9

Cumulated cluster frequency


0.8
0.7
0.6 Biopsies. Högberg (2015b)
0.5 Theory. Högberg (2015b)
0.4 0.64–0.36 Walrand et al. (2014)
0.3 0.50–0.50 Walrand et al. (2014)

0.2
0.1
0.0
0 10 20 30 40 50 60 70 80 90 100
Microspheres number per cluster

Figure 9.10 Cumulated cluster size distribution in biopsies of normal liver tissue explanted 9 days
after radioembolization using resin microspheres: observed (diamonds) and predicted (straight line).
(Derived from Figure 12A in Högberg, J., Small-Scale Absorbed Dose Modelling in Selective Internal
Radiation Therapy: Microsphere Distribution in Normal Liver Tissue, MA: University of Gothenburg,
2015b). The mean microsphere number per triad is 9.2. Dotted and dashed lines: Predictions using the
model of Walrand et al. (2014a) with a microsphere partition probability of 50%–50% and 64%–36% at
the bifurcation nodes, respectively.

A good agreement was obtained for the cumulated first process is completely deterministic and mono-
cluster size distribution (straight line in Figure tonically dependent on the absorbed dose according
9.10) and for the cluster frequency in the different to the well-known relation (Barendsen, 1962):
artery generations as well (Figure 12A in Högberg,
2015b). Currently, this arterial tree model is in the SF =  e−αD −βD
2
(9.8)
form of a schematic two-dimensional (2D) arbo-
rescence. Additional assumptions on the spatial where SF is the survival fraction, α and β are the
distribution of the clusters are needed in order to linear and quadratic radiosensitivities, and D is
compute the absorbed dose distribution. the absorbed dose (assumed to be instantaneously
delivered).
Organ recovery is characterized by a dose
9.12 MICrOSPHErE threshold that is tissue dependent. However, this
threshold is also variable among individuals of
DIStrIBUtION aND
the same species due to genetic differences and
HEPatIC tOXICItY also variations in metabolism between individu-
als. Therefore, organ recovery frequency as a func-
The first interesting quantitative result obtained tion of the absorbed does not exhibit a step shape,
from the microsphere transport simulations was to but rather a sharp sigmoid shape. This defines a
show that the typical therapy doses of 40 and 120 region around the dose threshold where the com-
Gy delivered to the liver by using resin and glass 90Y plete recovery displays some random nature. The
loaded microspheres, respectively, provide similar hepatic lobule is a functional tissue subunit act-
dose distribution to the portal triad—the critical ing as an independent organ on its own; therefore,
radiosensitive structure in liver radioembolization the recovery of a population of lobules can thus be
(Walrand et al., 2014a). described by a sigmoid function.
After tissue irradiation, two processes occur: a In science, we strive to describe the behavior
fraction of cells are killed, followed by either a com- of a large set of observations by a single formal-
plete recovery or loss of the tissue. Due to the huge ism or theory. For example, in the present case, we
number of cells and of electron tracks involved, the aim to develop a formalism to describe the hepatic
212 Microsphere deposition, dosimetry, radiobiology at the cell-scale

toxicity observed in EBRT and that observed in Equation 9.2 with the glass microsphere distribu-
liver radioembolization. The two modalities are tion obtained using the microsphere transport MC
characterized by a different dose rate: instanta- simulations corresponding to different BED deliv-
neous 1.5–2 Gy doses spaced in time (>8 hours) ered to the liver parenchyma. The best agreement
in EBRT and exponentially decreasing dose rate with studies in glass and resin microsphere radio-
in liver radioembolization (half-life = 2.7 days embolization (Chiesa et al., 2015; Strigari et al.,
for 90Y). The biological effective dose (BED) con- 2010, respectively) was obtained using the asym-
cept based on a linear-quadratic model (LQM) metric probability of 69%–31% for the microsphere
has been introduced to account for the dose rate bifurcation partition (Figure 9.11). This value is not
(Fowler, 1989). This concept succeeded in unifying far from the 64% to 36% value giving the best agree-
the renal toxicity observed in EBRT and in peptide ment with the cluster size distribution observed
receptor radionuclide therapy (PRRT) (Barone (Högberg, 2015). Note that a misunderstanding
et al., 2005; Wessels et al., 2008) and is now also occurred in Walrand et al. (2014b) where for the
considered in liver radioembolization (Cremonesi MC simulations the median toxic dose (TD50)
et al., 2008; Strigari et al., 2010). Regarding the was given in the targeted liver region as done in
irradiation itself, both the photons used in EBRT EBRT, while clinical TD50 data observed in radio-
and the β-particles used in liver radioemboliza- embolization were reported averaged on the whole
tion are low linear energy transfer (LET) particles liver (Chiesa et al., 2015; Strigari et al., 2010). This
and thus share the same radiobiology effectiveness explains why 60%–40% was the previous optimal
(RBE) (ICRP, 2007). microsphere partition probability.
Partial liver irradiation in EBRT (Dawson et al. An important benefit of the MC simulation is
2001) showed that NTCP = 0.5 is obtained by kill- the prediction of the hepatic toxicity as a func-
ing ≈40% of the liver volume or by irradiating 100%, tion of various parameters. Figure 9.12 shows the
80%, or 66% of the liver volume with a BED of 77, 95, WLTD50, i.e., the dose averaged over the whole
or 115 Gy, respectively. These results can be described liver giving NTCP = 0.5, as a function of the
by the following the sigmoid curve for the lobule targeted liver fraction and of the microsphere-
nonrecovery probability (Walrand et al. 2014b): specific activity (msA). For resin microspheres,
1 the WLTD50 is almost constant for a targeted liver
NR ( bed ) =   2.12
(9.9) fraction larger than 65%, reducing the drawback of
 93.8 
1+  mixing whole and right liver radioembolizations
 bed 
in the same NTCP reporting (Strigari et al., 2010).
The liver NTCP as a function of the killed lobule Note that Equations 9.9 and 9.10 derived from
fraction (KF) is (Dawson et al. 2001) EBRT involve that the WLTD50 become infinite
1 when the targeted liver fraction is lower than 40%.
NTCP (KF ) =   8.29
 0.4  (9.10)
1+  
 KF 
where KF can be computed using
9.13 aPPLICatION OF tHE
HEPatIC tOXICItY MODEL
KF =   ∑ν (bed ) NR(bed )
i
i i (9.11)
For convenient use, the WLTD(p,Vf,msA), i.e., the
whole-liver dose providing a NTCP = p when tar-
where ν(BEDi) is the fraction of lobules receiv-
geting a liver volume fraction Vf using 90Y loaded
ing a dose BEDi. Note that in EBRT, Equation 9.11
microspheres of specific activity msA, is fitted by
reduces to
WLTD( p,Vf ,msA ) = 47.1 Gy   
KF = Vf  NR(BED) (9.12)
×    
(1 + 0.457 p )    F (msA)  Vf   (9.13)
where Vf is the irradiated liver fraction.
Walrand et al. (2014b) computed the ν(BEDi)
(Vf − Kf ( p)) 0.869 F (msA)
fractions by convolving the Russell dose kernel where the dimensionless scale factor F(msA) is
9.13 Application of the hepatic toxicity model 213

SPECT–based VOI drawing method


1.2

0.8

0.6
NTCP

0.4
MC simulation

0.2
NTCP (D)
FIT NTCP (D)
0 95% CI

–0.2
0 20 40 60 80 100 120 140 160 180 200
D [Gy]
(a)

Liver D (Gy)
0 35 55 72 87
1.0
0.8
Liver NTCP
0.4 0.6

MC simulation
0.2

Model
95% CI
Exp
0.0

0 50 100 150 200


Liver BED (Gy)
(b)

Figure 9.11 NCTP comparison between clinical observations and prediction (squares) from Equations
9.9 and 9.10 using 69%–31% as the microsphere partition probability in the MC calculation of ν(BEDi).
(a) Glass microspheres in right liver radioembolizations (Vf ≈ 0.66) (With kind permission from
Springer Science + Business Media: Eur J Nucl Med Mol Imaging, Radioembolization of hepatocarci-
noma with (90)Y glass microspheres: Development of an individualized treatment planning strategy
based on dosimetry and radiobiology, 42, 2015, 1718–1738, Chiesa et al.). (b) Resin microspheres in
mixed whole and right liver radioembolizations (Vf ≈ 0.80). (Reprinted from Strigari et al., J Nucl Med,
51, 1377–1385, 2010. With permission of the Nuclear Medicine Society). Squares were added by the
author. Doses are averaged over the whole liver parenchyma.
214 Microsphere deposition, dosimetry, radiobiology at the cell-scale

200

180

ms A
160
2.5
WLTD50 [Gy]
140 kB
0.9 q
4k
Bq 0.94 kBq: Chiesa et al. 2015
120 0.4
5k
Bq
100
0.15
kBq
80

60 0.05 kBq
0.05 kBq: Strigari et al. 2010

40
0.5 0.6 0.7 0.8 0.9 1
Targeted liver fraction
(a)

120 Vf = 0.66

110

Vf = 0.8
100

90 Vf = 1.0
WLTD50 [Gy]

80

70

60

50

40
0.00 0.50 1.00 1.50 2.00 2.50
Microsphere specific activity (kBq)
(b)

Figure 9.12 Circles: WLTD50 (i.e., dose averaged on the whole liver parenchyma giving NTCP = 0.5)
derived from Equations 9.9 and 9.10 using a 69%–31% microsphere partition probability in the MC
simulation of ν(BEDi ). Straight lines: fit with Equations 9.13 and 9.14. (a) WLTD50 as a function of the
targeted liver fraction for different microsphere-specific activities (msA). (b) WLTD50 as a function of
the msA for different targeted liver volume fractions (Vf ).

F ( msA ) =   1 − e ( − 3 msA/0.0471 kBq


) (9.14)
And Kf(p) is the inverse of the relation
(Equation 9.10), i.e.,
Note that, for a fixed total radioembolization p
activity, the cube root in F(msA) is a quantity Kf ( p) =  0.4 8.29 (9.15)
1− p
strongly proportional to the mean intermicro-
sphere distance in the liver.
References 215

Kf(0.5) = 0.4 and Kf(0.05) = 0.28, which means reconcile the liver toxicities observed in EBRT
that ablating by irradiation 40% and 28% of the and in radioembolization using glass and resin
liver induces a NTCP of 50% and 5%, respectively, microspheres. More interestingly, these MC sim-
as observed in EBRT (Dawson et al., 2001). ulations can be fitted with an analytical formula
In the fitted domain {msA є (0.05,0.15, (Equations 9.13 and 9.14) that allows performing
0.45,0.94,2.5) kBq, p є (0.15,0.30,0.50), Vf є of individualized radioembolization planning as
(0.55,0.66,0.80,1.00)} the mean absolute relative a function of the liver fraction targeted and of the
deviation of the fit versus MC simulations (i.e., mSA used.
fit − MC was 2.9% with extreme relative devia- This model contains an adjustable param-
 )
MC eter, i.e., the microsphere partition probability
tions (i.e., fit − MC )  of –4.97% and 4.60%. It is at the arterial nodes, which was fitted in order to
MC reproduce the liver toxicities observed in clinical
quite remarkable that such a simple expression of radioembolization studies (Strigari et al., 2010;
F accurately takes into account the impact of mSA. Chiesa et al., 2015). However, the obtained parti-
As the microspheres are localized in the por- tion probability 69%–31%: (1) is within the range of
tal triads, the WLTD(p,Vf,msA) value when msA probabilities predicted by microsphere transport
vanishes does not approach that of EBRT. Indeed, dynamic simulations (Kennedy et al., 2010), (2) is
TD50 = 43 Gy is the dose of 90Y corresponding to close to the value 64%–36% fitting the observed
BED50 = 77 Gy observed in whole-liver EBRT. The microsphere cluster distribution (Högberg et al.,
simulation in Table 9.1 shows that, in the case of trans- 2015), (3) fits both the hepatic toxicity reported in
lation invariant microsphere distribution (which is glass and resin microsphere radioembolization,
expected when the microsphere number increases), 43 and (4) unifies hepatic toxicity observed in radio-
Gy in the triad arteries corresponds to a liver paren- embolization and in EBRT. These four facts sup-
chyma dose of 40 × 43/118 = 15 Gy, i.e., BED = 19 Gy, port the coherence of the whole model and also of
fourfold smaller than the 77 Gy observed in EBRT. the reported clinical hepatic toxicity as well.
As there is no clinical study available in order to
validate the MC simulation for microsphere activ-
ity below 0.05 kBq, a conservative expression of
REFERENCES
F(msA) was chosen such as
lim WLTD ( p,Vf ,msA ) = 0 (9.16) Barendsen, G.W. (1962). Dose-survival curves of
msA →0 human cells in tissue culture irradiated with
Equations 9.13 and 9.14 could be used to compute alpha-, beta-, 20-kV x- and 200-kV x-radiation.
the needed absorbed dose and also the activity in Nature 193:1153–1155.
order to achieve a desired NTCP = p as a function Barone, R. et al. (2005). Patient-specific dosim-
of the targeted liver fraction volume and mSA. Note etry in predicting renal toxicity with (90)
that this set of equations was obtained by tuning Y-DOTATOC: Relevance of kidney volume and
one parameter of the model (the microsphere parti- dose rate in finding a dose-effect relation-
tion probability at the arterial tree nodes) in order ship. J Nucl Med 46:99S–106S.
to fit the clinical hepatic toxicity observed in glass Basciano, C.A. (2010). Computational Particle-
(Chiesa et al., 2015) and in resin (Strigari et al., 2010) Hemodynamics Analysis Applied to an
radioembolization. Thus, the prediction accuracy of Abdominal Aortic Aneurysm with Thrombus
Equations 9.13 and 9.14 does not only depend on the and Microsphere-Targeting of Liver Tumors.
goodness of the model but also on the goodness of Raleigh, NC: North Carolina State University.
the observed clinical hepatic toxicity. Campbell, A.M., Bailey, I.H., Burton, M.A. (2000).
Analysis of the distribution of intra-arterial
microspheres in human liver following hepatic
yttrium-90 microsphere therapy. Phys Med
9.14 CONCLUSIONS Biol 45:1023–1033.
Chiesa, C. et al. (2011). Need, feasibility and con-
MC simulations, using a very simple model of the venience of dosimetric treatment planning in
hepatic arterial tree and microsphere transport, liver selective internal radiation therapy with
216 Microsphere deposition, dosimetry, radiobiology at the cell-scale

(90)Y microspheres: The experience of the Högberg, J. et al. (2014). Heterogeneity of


National Tumor Institute of Milan. Q J Nucl microsphere distribution in resected liver and
Med Mol Imaging 55:168–197. tumour tissue following selective intrahepatic
Chiesa, C. et al. (2015). Radioembolization of radiotherapy. EJNMMI Res 4:48.
hepatocarcinoma with (90)Y glass micro- Högberg, J. et al. (2015a). Increased absorbed
spheres: Development of an individualized liver dose in Selective Internal Radiation
treatment planning strategy based on dosim- Therapy (SIRT) correlates with increased
etry and radiobiology. Eur J Nucl Med Mol sphere-cluster frequency and absorbed dose
Imaging 42:1718–1738. inhomogeneity. EJNMMI Phys 2:10.
Cnuddea, V., Boone, M. (2013). High-resolution Högberg, J. (2015b). Small-Scale Absorbed Dose
X-ray computed tomography in geosciences: Modelling in Selective Internal Radiation
A review of the current technology and appli- Therapy: Microsphere Distribution in Normal
cations. Earth Sci Rev 123:1–17. Liver Tissue. Gothenburg: University of
Crawford, A.R., Lin, X.Z., Crawford, J.M. (1998). Gothenburg.
The normal adult human liver biopsy: A ICRP. (2007). The 2007 recommendations of the
quantitative reference standard. Hepatology international commission on radiological
28:323–331. protection. ICRP publication 103. Ann ICRP
Cremonesi, M. et al. (2008). Radioembolisation 37:1–332.
with 90Y-microspheres: Dosimetric and Kennedy, A.S. et al. (2004). Pathologic response
radiobiological investigation for multi-cycle and microdosimetry of (90)Y microspheres in
treatment. Eur J Nucl Med Mol Imaging man: Review of four explanted whole livers.
35:2088–2096. Int J Radiat Oncol Biol Phys 60:1552–1563.
Cross, W.G., Freedman, N.O., Wong, P.Y. (1992). Kennedy, A. et al. (2007). Recommendations
Tables of Beta-Ray Dose Distributions in for radioembolization of hepatic malignan-
Water. Report no. AECL-10521. Ontario, cies using yttrium-90 microsphere brachy-
Canada: Atomic Energy of Canada, Ltd. therapy: A consensus panel report from the
Dawson, L.A., Ten Haken, R.K., Lawrence, T.S. radioembolization brachytherapy oncology
(2001). Partial irradiation of the liver. Semin consortium. Int J Radiat Oncol Biol Phys
Radiat Oncol 11:240–246. 68:13–23.
Debbaut, C. et al. (2012). Perfusion character- Kennedy, A.S., Kleinstreuer, C., Basciano, C.A.,
istics of the human hepatic microcirculation Dezarn, W.A. (2010). Computer model-
based on three-dimensional reconstructions ing of yttrium-90-microsphere transport in
and computational fluid dynamic analysis. the hepatic arterial tree to improve clini-
J Biomech Eng 134:011003. cal outcomes. Int J Radiat Oncol Biol Phys
Debbaut, C. et al. (2014). Analyzing the human 76:631–637.
liver vascular architecture by combining Lau, W.Y. et al. (2012). Patient selection and
vascular corrosion casting and micro-CT scan- activity planning guide for selective inter-
ning: A feasibility study. J Anat 224:509–517. nal radiotherapy with yttrium-90 resin
Fowler, J.F. (1989). A review: The linear quadratic microspheres. Int J Radiat Oncol Biol Phys
formula and progress in fractionated radio- 82:401–407.
therapy. Br J Radiol 62:679–675. Ljungberg, M., Strand, S.-E., King, M.A. (2013).
Gulec, S.A. et al. (2010). Hepatic structural dosim- Monte Carlo Calculations in Nuclear Medicine.
etry in 90Y microsphere treatment: A Monte Applications in Diagnostic Imaging. Boca
Carlo modeling approach based on lobular Raton, FL: CRC Press.
microanatomy. J Nucl Med 51:301–310. Mettler, F.A., Guiberteau, M.J. (2012). Essentials
Haga, J. et al. (2008). Liver regeneration in of Nuclear Medicine Imaging. Philadelphia,
donors and adult recipients after living PA: Saunders.
donor liver transplantation. Liver Transpl Michalopoulos, G.K., DeFrances, M.C. (1997).
14:1718–1724. Liver regeneration. Science 276:60–66.
References 217

Paxton, A.B., Davis, S.D., Dewerd, L.A. (2012). Keller A, Heinrich S, Niederreiter H (Eds.)
Determining the effects of microsphere and Monte Carlo and Quasi-Monte Carlo Methods
surrounding material composition on 90Y 2006. Berlin: Springer-Verlag.
dose kernels using egsnrc and mcnp5. Med Schreiner, W., Buxbaum, P.F. (1993). Computer-
Phys 39:1424–1434. optimization of vascular trees. IEEE Trans
Pillai, K.M. et al. (1991 ). Microscopic analysis of Biomed Eng 40:482–491.
arterial microsphere distribution in rabbit liver Schroeder, L. (1974). Buffon’s needle problem:
and hepatic VX2 tumor. Selective Cancer Ther An exciting application of many mathematical
7:39–48. concepts. Mathematics Teacher 67:183–186.
Press, W.H., Teukolsky, S.A., Vetterling, W.T., Schwen, L.O., Preusser, T. (2012). Analysis
Flannery, B.P. (2007). Numerical Recipes in C: and algorithmic generation of hepatic
The Art of Scientific Computing. Cambridge, vascular systems. Int J Hepatol.
England: Cambridge Press. doi:10.1155/2012/357687
Richards, A.L. et al. (2012). Experimental micro- Strigari, L. et al. (2010). Efficacy and toxicity
sphere targeting in a representative hepatic related to treatment of hepatocellular car-
artery system. IEEE Trans Biomed Eng cinoma with 90Y-SIR spheres: Radiobiologic
59:198–204. considerations. J Nucl Med 51:1377–1385.
Richards, A.L. (2013). Experimental Investigations Walrand, S. et al. (2014a). The low hepatic toxic-
Into the Targeted Delivery of Microspheres in ity per Gray of 90Y glass microspheres is
Radioembolization Therapy. PhD dissertation. linked to their transport in the arterial tree
Raleigh, NC: North Carolina State University, favoring a nonuniform trapping as observed
191 pp. in posttherapy PET imaging. J Nucl Med
Roberson, P.L. et al. (1992). Three-dimensional 5:135–140.
tumor dosimetry for hepatic yttrium- Walrand, S., Hesse, M., Jamar, F., Lhommel, R.
90-microsphere therapy. J Nucl Med Official (2014b). A hepatic dose-toxicity model open-
Publ 33:735–738. ing the way toward individualized radioembo-
Russell, J.L., Carden, J.L., Herron, H.L. (1988). lization planning. J Nucl Med 55:1317–1322.
Dosimetry calculation for yttrium-90 used in Wessels, B.W. et al. (2008). MIRD pamphlet No.
the treatment of liver cancer. Endocuriether 20: The effect of model assumptions on
Hyperther Oncol 4:171–186. kidney dosimetry and response—Implications
Saito, M., Matsumoto, M. (2008). SIMD ori- for radionuclide therapy. Nucl Med
ented fast Mersenne twister: A 128-bit 49:1884–1899.
pseudorandom number generator. In
PART 4
Following Patients Treated
with Radioembolization

10 Postradioembolization imaging using bremsstrahlung 90Y SPECT/CT 221


S. Cheenu Kappadath
11 Quantitative postradioembolization imaging using PET/CT 229
Marco D’arienzo, Luca Filippi, and Oreste Bagni
12 Image-based three-dimensional dosimetry following radioembolization 251
Alexander S. Pasciak and S. Cheenu Kappadath
13 Diagnostic reporting using postradioembolization imaging 263
Yung Hsiang Kao
14 The use of postprocedural imaging in the medical management of patients 281
Austin C. Bourgeois, Marcelo S. Guimaraes, Yong C. Bradley,
Christopher Hannegan, and Alexander S. Pasciak
10
Postradioembolization imaging using
bremsstrahlung 90Y SPECT/CT

S. CHEENU KAPPADATH

10.1 Background and rationale 221 10.3.1 Planar imaging 222


10.2 Challenges associated with 10.3.2 SPECT and SPECT/CT imaging 223
90Y SPECT imaging 222 10.4 Summary and conclusions 225
10.3 Approaches to 90Y bremsstrahlung References 226
imaging 222

2010). Overall accuracies of 72%, 79%, and 96% for


10.1 BaCKGrOUND aND planar, SPECT, and SPECT/CT, respectively, have
ratIONaLE been reported (Hamami et al., 2009).
There are a number of qualitative studies that
90Y radioembolization is used in the management of have suggested that MAA distributions observed
patients with unresectable primary and metastatic during planning often match the 90Y microsphere
liver cancers. 99mTechnetium-labeled macroaggre- distributions after therapy. Concordance between
gated albumin (99mTc MAA) has been successfully MAA and 90Y has been reported (Chiesa et al., 2015)
used, at least in terms of patient safety, as a surrogate that increases in confidence when selective segmen-
radiopharmaceutical for treatment planning since tal or lobar therapies are planned (Kao et al., 2013).
the inception of the therapy, as described in Chapter However, as reviewed in Chapter 4, several studies
4. Planar scintigraphy of 99mTc MAA has been used have also shown that the distribution of MAA dur-
primarily to evaluate the lung shunt fraction and ing treatment planning may not be a consistent and
estimate the mean absorbed dose to lung after the 90Y reliable indicator of the distribution of the 90Y micro-
radioembolization treatment (Ho et al., 1997). MAA spheres after the administration of treatment (Ilhan
uptake in the lung consequently affects the prescrip- et al., 2015). Differences of greater than 20% uptake
tion of administered 90Y microsphere activity to pre- between 99mTc MAA and 90Y have been reported in
vent radiation pneumonitis. MAA distribution in 43% (97/225) of cases (Wondergem et al., 2013).
vivo is also used to assess extrahepatic distribution Furthermore, especially in the past decade, in
and judge the adequateness of tumor perfusion from vivo 99mTc MAA distributions have begun to be
the catheter placement. It has been demonstrated used in conjunction with dosimetry models such as
that the assessment of MAA distribution with single- the medical internal radiation dose (MIRD) (Gulec
photon emission computed tomography/CT (SPECT/ et al., 2006) or partition (Ho et al., 1996) models
CT) is superior compared with SPECT, which in turn to calculate the tumor and normal liver doses (see,
is superior to planar imaging (Ahmadzadehfar et al., e.g., Chiesa et al., 2015; Garin et al., 2013).

221
222 Postradioembolization imaging using bremsstrahlung 90Y SPECT/CT

However, the potential discrepancies in distri- of the continuous nature of the energy spectrum
bution between planning 99mTc MAA and treat- and the lack of readily identifiable spectral charac-
ment 90Y radioembolization argue for the need for teristics, which result in high levels of scatter. The
posttreatment 90Y imaging to assess the delivered lack of photo-peak emissions has been a major bar-
distribution of 90Y treatment. Investigations have rier for the standardization of a 90Y bremsstrah-
demonstrated the positive role of 90Y planar and 90Y lung imaging procedure; consequently, there is a
bremsstrahlung SPECT/CT in the management of wide variability in image quality among different
patients after radioembolization (Ahmadzadehfar facilities.
et al., 2012). 90Y SPECT/CT with an overall accu- Historically, only planar imaging of the 90Y
racy of 99% was shown to be superior to SPECT bremsstrahlung emission, if at all, was performed
and planar imaging in the prediction of gastroin- after 90Y therapies to confirm the 90Y microsphere
testinal (GI) ulcers. distribution within the liver compartment; how-
In addition, 90Y SPECT/CT is also being ever, the nonuniform uptake of microspheres
investigated for the calculation of tumor and in the liver cannot be visualized because of poor
normal liver doses using the MIRD or partition image contrast. 90Y bremsstrahlung SPECT, if per-
dosimetry models, as described in Chapter 5. formed, used a wide energy window and recon-
Furthermore, as described in Chapter 12, quan- structed using filtered back-projection (FBP)
titative 90Y SPECT/CT also facilitates dosimetry with no compensations for scatter or attenuation.
at the voxel level that allows for investigations Therefore, historically, 90Y-SPECT images have
into the volume distribution of absorbed doses in been nonquantitative and have poor contrast and
tumors and normal liver (Kappadath et al., 2014). resolution.
The role of postradioembolization imaging,
whether it is performed with 90Y SPECT/CT or
90Y PET/CT, is absolutely critical when it comes
10.3 aPPrOaCHES tO 90Y
to assessing tumor dose–response relationships
BrEMSStraHLUNG
and radioembolization-induced liver disease.
The role of 90Y PET/CT is reviewed in Chapter 11.
IMaGING
However, this chapterwill focus on the technical
aspects related to 90Y bremsstrahlung SPECT/CT The technical approaches for improving 90Y
for posttreatment imaging. bremsstrahlung imaging have included evalu-
ation using phantoms, simulations, and patient
data. The major parameters investigated with
respect to data acquisition have been energy win-
10.2 CHaLLENGES aSSOCIatED dow selection for imaging, and choice of colli-
WItH 90Y SPECt IMaGING mation. The major parameters investigated with
respect to data processing for SPECT imaging
As discussed in Chapter 1, 90Y decays via beta have been correction techniques for scatter and
emission to 90Zr with a half-life of 64.1 hours and attenuation, iterative reconstructions, partial
with a maximum beta energy of 2.28 MeV. In con- volume correction, and calibration for quantita-
trast with the majority of radionuclides used in tive SPECT. There has also been some early work
nuclear medicine imaging, 90Y is effectively a pure done on image filtration for noise suppression in
beta emitter, i.e., it lacks discrete energy photon planar images.
emissions, such as gamma and/or characteristic
fluorescence X-rays; therefore, imaging of 90Y with 10.3.1 PLANAR IMAGING
gamma cameras is challenging.
90Y activity distribution in vivo is traditionally The 90Y bremsstrahlung planar acquisition para-
assessed by imaging the bremsstrahlung photon— meters of collimator, sensitivity, and energy
produced from interactions of the energetic beta window have been empirically evaluated using
particles with soft tissue—using a gamma camera phantoms. The spectral components of 90Y brems-
or by SPECT/CT. Bremsstrahlung photons are not strahlung images have been investigated and
considered to be well suited for imaging because improved spatial resolution and image contrast
10.3 Approaches to 90Y bremsstrahlung imaging / 10.3.2 SPECT and SPECT/CT imaging 223

reported for 90Y bremsstrahlung imaging with a et al. (1994a). These studies also showed that a
medium-energy (ME) collimator when compared wide energy range of 50–200 keV could increase
with a low-energy collimator with an energy the sensitivity without substantial loss of spatial
window of 55–285 keV (Shen et al., 1994a). The resolution or contrast.
marginal improvement in resolution observed Simulation studies have shown that 90Y brems-
with the high-energy collimator was offset by the strahlung emission spectrum from the liver can
large decrease in its sensitivity. Early attempts at be considered to be a summation of the following
quantitative 90Y planar imaging have focused on spectral components: primary bremsstrahlung,
image restoration using filtration; both Wiener object scatter, camera backscatter, collimator
(King et al., 1983; Shen et al., 1994b) and wavelet- scatter and penetration, and lead X-rays from the
based neural network (Qian et al., 1994b) filters collimator. The photons within the energy spec-
have been studied. These approaches use image trum between 70 and 100 keV have large con-
filtration to deconvolve scatter and septal pen- tributions of characteristic X-rays originating
etration and have shown improvement in energy primarily from the lead collimator. The energy
resolution and image contrast. Filtered brems- spectrum between 200 and 300 keV was shown
strahlung geometric-mean images (that partially to predominantly arise from backscatter, while
compensate for attenuation) yielded individ- photons higher than 300 keV were from collima-
ual activities within 17% (Shen et al., 1994a). tor scatter and septal penetration. At any given
However, filter-based deconvolutions are image energy window, the ratio of primary bremsstrah-
dependent and require phantom calibration for lung to the total photons detected is typically
quantification (Shen et al., 1994b). In addition, less than 15%, with the highest primary fraction
they can result in image artifacts and overcom- around 80–180 keV (Heard et al., 2004). Some
pensation of the system response functions (King previous studies have suggested using acquisi-
et al., 1991). The superpositioning of overlapping tion energy windows with a lower threshold than
90Y sources (tumor and normal liver) inherent in 100 keV because of the large number of detected
planar images outweigh their quantitative capa- photons (Shen et al., 1994; Heard et al., 2004).
bilities; therefore, quantitative planar techniques However, this appears to be not a good choice for
have not been applied to clinical 90Y imaging accurate quantitative imaging unless the high-
studies. order scatter can be modeled accurately in the
projector (Rong et al., 2012a).
10.3.2 SPECT AND SPECT/CT Other studies have also reported on 90Y brems-
IMAGING strahlung SPECT imaging using phantoms.
Minarik et al. (2008) evaluated 90Y bremsstrah-
Historically, only ad hoc arguments about the lung SPECT for 90Y ibritumomab tiuxetan with
various kinds of photons have been proposed a General Electric VH/Hawkeye system using
to optimize the acquisition energy window for CT-based attenuation correction, scatter-kernel-
90Y bremsstrahlung imaging. Recent improve- based scatter correction, model-based collimator
ments in computational speed and process- response, and iterative reconstruction. SPECT
ing power have enabled Monte Carlo (MC) data of an anthropomorphic torso phantom
simulation-based investigations of 90Y brems- with liver insert was acquired over a relatively
strahlung spectral decomposition to elucidate wide 105–195 keV energy range and reconstruc-
the spectral composition of 90Y bremsstrahlung tion accuracies of 10%–16% were reported. They
(see e.g., Heard et al., 2004; Minarik et al., 2008; substantiate expectations that phantom-based
Rault et al., 2010; Elschot et al., 2013). Heard et al. sensitivity calibration can yield accurate quan-
(2004) used the EGSnrc MC code to simulate the tification of 90Y SPECT images. However, they
Philips/ADAC Forte gamma camera, and Rault reported results for only one phantom experi-
et al. (2010) used the Geant4/GATE MC to simu- ment and the scatter kernel was computed only
late the Philips AXIS/IRIS gamma camera. These for one configuration. Their use of a single linear-
studies corroborate that ME collimators and a attenuation coefficient for CT-based attenuation
100–150 keV energy window provided optimal correction over a wide 105–195 keV energy win-
image contrast, as previously suggested by Shen dow could, however, potentially introduce errors
224 Postradioembolization imaging using bremsstrahlung 90Y SPECT/CT

from the large variation of linear attenuation method achieved excellent accuracy in model-
across the wide energy window. ing photon counts (errors ~1%) and the quantita-
Rong and Frey published a series of papers that tive accuracy of activity estimates for all organs
critically investigated energy windows for imag- (errors were below ~12%). The net percent errors
ing 90Y bremsstrahlung SPECT/CT (Rong et  al., in activity estimates from physical geometrical
2012a, 2012b). Rong et al. (2012a) developed an phantom experiments were shown to be 7%–10%,
optimization method that takes into account and in the simulated patient data corresponded
both the bias and the variance of the activity to mean organ dose errors of ~10% for the lung
estimates for optimizing acquisition energy win- and 4%–5% for the liver.
dow for quantitative 90Y bremsstrahlung SPECT In a different technique, the Utrecht Monte
imaging. They used the weighted root mean Carlo Simulator was adapted for 90Y and incorpo-
squared error of volume of interest (VOI) activ- rated into a statistical reconstruction algorithm
ity estimates, which took into account both the by Elschot et al. (2013). Photon scatter and attenu-
bias due to model mismatch and the variance of ation for all photons sampled from the full 90Y
the VOI activity estimates, as the figure of merit energy spectrum were modeled during reconstruc-
for optimization. They concluded that the opti- tion by Monte Carlo simulations. The energy- and
mal acquisition energy window for 90Y SPECT distance-dependent collimator–detector response
imaging was 100–160 keV. To obtain the optimal was modeled with precalculated convolution ker-
acquisition energy window for general situations nels. For the purpose of computationally efficient
of interest in clinical 90Y microsphere imaging, modeling of the distance- and energy-dependent
they generated phantoms with multiple tumors collimator-detector response (CDR), the updated
of various sizes and various tumor-to-normal photon intensities were binned voxel-wise in eight
activity concentration ratios using a digital phan- energy-dependent, three-dimensional (3D) scatter
tom that realistically simulates human anatomy. maps according to their energy after the last scat-
90Y microsphere imaging was then simulated ter event; the energy of photons ranged from 50 to
with a clinical SPECT system and typical imag- 2000 keV. The energy used for the generation of the
ing parameters using a previously validated MC final image was 50–250 keV. The quantitative accu-
code and a previously proposed method for mod- racy of 90Y bremsstrahlung SPECT was substan-
eling the image degrading effects in quantitative tially improved by Monte Carlo-based modeling
SPECT reconstruction. of the image degrading factors. The International
In another study, Rong et al. (2012b) used Electrotechnical Commission (IEC) image qual-
simulations of imaging nuclear detectors ity phantom was used to quantitatively evaluate
(SIMIND) to accurately model image degrad- the performance of their approach in comparison
ing factors such as object attenuation, scat- with those of clinical SPECT reconstruction. Their
ter, and the collimator–detector response, all approach demonstrated substantially improved
of which are energy dependent but essential to image contrast in patient scans from 25% to 88% for
obtain quantitatively accurate images. Their the 37 mm sphere and decreased the mean residual
approach used a single, wide acquisition win- count error in the lung insert from 73% to 15% at the
dow (100–500 keV), with separate treatment of cost of higher image noise.
photons in various energy ranges and in various While there have been a number of studies to
logical categories during the modeling process. improve 90Y bremsstrahlung imaging both quali-
In particular, they separated photons into eight tatively and quantitatively, most of the published
categories based on energy and logical category. approaches require some sort of Monte Carlo sim-
Primary and scattered photons (i.e., photons not ulation, which cannot be easily implemented in
scattered in the body) were separated into four routine clinical practice. This has led some groups
categories according to their emission energies: to develop a simple 90Y bremsstrahlung SPECT/
0–250, 250–500, 500–1000, and 1000–2000 keV. CT imaging protocol based on energy-window-
They demonstrated a good agreement between based scatter compensation and CT-based atten-
the experimental measurement and Monte uation correction that is readily implemented in
Carlo simulation. In the extended cardiac-torso commercial SPECT/CT systems, yet improves
(XCAT) phantom simulation, the proposed 90Y bremsstrahlung SPECT/CT image quality
10.4 Summary and conclusions 225

and quantification (Siman et al., 2016). Based on energy window ranging from 50 to 350 keV. By
phantom experiments, subsequently verified on using the recorded event energy in the recon-
patient images, they determined that energy win- struction method, shorter acquisition time and
dows 90–125 keV and 310–410 keV were suitable reduced orbit range will be feasible allowing the
imaging and scatter-estimate energy windows, design of a simplified mobile gantry.
respectively. For clinical images, the scatter cor-
rection factor was determined to be 0.5–0.6 with
a coefficient of variation of ~10%; hence using a
single-scatter correction factor was considered
10.4 SUMMarY aND
adequate. They acknowledge that their proposed CONCLUSIONS
energy-window-based scatter-correction method
only partially corrects for backscatter, septal There have been a lot of advances toward improv-
penetration, and septal scatter components, a ing imaging of 90Y bremsstrahlung emissions
claim supported by the simulation studies for using SPECT/CT (Figure 10.1). The general con-
components that dominate the scatter-estimate sensus appears to be that an imaging window
energy window. around 90–160 keV is suitable for qualitative
SPECT calibration factor is defined as the SPECT/CT imaging using ME collimators (Table
ratio of the total activity in the field of view 10.1). Quantitative SPECT/CT is also feasible
(FOV) to the total counts in the FOV. For ideal when additional corrections such as attenuation,
image reconstruction (with accurate corrections scatter, and collimator-response modeling are
for scatter, attenuation, and collimator–detector also incorporated during reconstruction. In fact,
response), calibration with a point source in when energy-dependent corrections are incorpo-
air will suffice. In practice, however, the image rated the energy window width can be expanded
reconstruction is not perfect; therefore, the to around 250 keV.
calibration factor is usually derived from phan- The potential for discrepancies in distribu-
tom images with all the necessary corrections tion between planning 99mTc MAA and treat-
applied. Posttherapy 90Y SPECT/CT scan pres- ment 90Y radioembolization strongly argues for
ents a unique condition where the total 90Y activ- the need for posttreatment 90Y imaging to assess
ity inside the liver (and hence inside the SPECT the delivered distribution of 90Y treatment. The
FOV) can be determined with uncertainty <10%. investigations summarized in this chapter read-
Clinical images acquired under such conditions ily facilitate the qualitative assessment of post-
can be used to calibrate the SPECT/CT imag- therapy 90Y microsphere distributions. In fact,
ing system. In clinical studies, the total SPECT quantitative 90Y SPECT/CT can also be achieved
counts observed were found to be proportional to without too much effort using commercial
the 90Y activity in the FOV. SPECT images with SPECT/CT scanners. Quantitative 90Y SPECT/
CT attenuation correction and scatter correc- CT facilitates dosimetry at the voxel level that
tion can be used to accurately quantify the activ- allows for investigations into the volume distri-
ity present in the FOV with an average absolute bution of absorbed doses in tumors and normal
deviation ≤5% (Siman et al., 2016). liver (see Chapter 12).
There have also been some very innovative The advancement of 90Y radioembolization
approaches into specifically imaging 90Y using from what is usually considered to be palliative
nonclinical gamma cameras. Walrand et al. into a more frontline therapy will rest heavily
(2014) used MC simulations of energy spectra and on the ability to determine the absorbed doses
showed that a camera based on a 30-mm-thick to tumor necessary to elicit a response. Once the
bismuth germanium oxide (BGO) crystal and tumor dose response following radioemboliza-
equipped with a high-energy pinhole collima- tion is known, 90Y therapy planning can be based
tor was well adapted to bremsstrahlung imag- on delivering tumoricidal doses. This advance-
ing. The total scatter contamination is reduced by ment in treatment planning will be essential in
a factor of 10 versus a conventional NaI camera the promotion of radioembolization. The ability
equipped with a high-energy parallel hole col- to accurately determine tumor doses stems from
limator, enabling acquisition using an extended the ability to quantify 90Y SPECT/CT, or by using
226 Postradioembolization imaging using bremsstrahlung 90Y SPECT/CT

(a) (b)

(c) (d) (e)

Figure 10.1 An illustration of different 90Y SPECT/CT image quality based on the data acquisition
and analysis schemas used. The fused SPECT/CT images (energy window 90–150) of the anthro-
pomorphic torso phantom through the same coronal slices showing the liver insert and two hot
spheres but with different SPECT reconstruction schemes: (a) filter backprojection (FBP), the
technique of choice when SPECT-only systems (i.e., no CT) are used; (b) ordered subsets maximum
likelihood expectation maximization (3D-OSEM) with neither attenuation nor scatter corrections;
(c) 3D-OSEM with only attenuation but no scatter correction; (d) 3D-OSEM with both attenuation
and scatter corrections; and (e) a photograph of the torso phantom. The series of images shows
the dramatic improvements in image quality achieved using 3D-OSEM iterative reconstruction
with both attenuation and scatter corrections; the background signal outside the liver is largely
accounted for while the spheres are visualized with better contrast. The SPECT display scale is
0%–60% of maximum SPECT counts.

other quantitative posttreatment imaging modal- REFERENCES


ities such as 90Y PET/CT, discussed in Chapter 11.
Ahmadzadehfar, H. et al. (2010). The signifi-
cance of 99mTc-MAA SPECT/CT liver per-
Table 10.1 Summary of energy window ranges fusion imaging in treatment planning for
for imaging 90Y bremsstrahlung emissions using 90Y-microsphere selective internal radiation
SPECT/CT together with the associated treatment. J Nucl Med 51:1206–1212.
publications Ahmadzadehfar, H. et al. (2012). The significance
of bremsstrahlung SPECT/CT after yttrium-90
Energy range (keV) references
radioembolization treatment in the prediction
105–195 Minarik et al. (2008) of extrahepatic side effects. Eur J Nucl Med
100–160 Rong et al. (2012a) Mol Imaging 39:309–315.
90–125 Siman et al. (2016) Chiesa, C. et al. (2015). Radioembolization of
100–500a Rong et al. (2012b) hepatocarcinoma with 90Y glass micro-
50–250a Elschot et al. (2013) spheres: Development of an individualized
a The energy ranges indicate that energy-dependent
treatment planning strategy based on dosim-
corrections are necessary when using those energy etry and radiobiology. Eur J Nucl Med Mol
windows. Imaging 42:1718–1738.
References 227

Elschot, M. et al. (2013). Quantitative Monte King, M.A., Doherty, P.W., Schwinger, R.B., Penney,
Carlo–based 90Y SPECT reconstruction. B.C. (1983). A Wiener filter for nuclear medicine
J Nucl Med 54:1557–1563. images. Med Phys 10(6):876–880.
Garin, E. et al. (2013). Boosted selective inter- Minarik, D., Gleisner, K.S., Ljungberg, M. (2008).
nal radiation therapy with 90Y-loaded glass Evaluation of quantitative 90Y SPECT based
microspheres (B-SIRT) for hepatocellular carci- on experimental phantom studies. Phys Med
noma patients: A new personalized promis- Biol 53:5689–5703.
ing concept. Eur J Nucl Med Mol Imaging Qian, W., Clarke, L.P. (1996). A restoration
40:1057–1068. algorithm for P-32 and Y-90 bremsstrah-
Gulec, S.A., Mesoloras, G., Stabin, M. (2006). lung emission nuclear imaging: A wave-
Dosimetric techniques in 90Y-microsphere let-neural network approach. Med Phys
therapy of liver cancer: The MIRD equations for 23(8):1309–1323.
dose calculations. J Nucl Med 47:1209–1211. Rault, E. et al. (2010). Fast simulation of
Hamami, M.E. et al. (2009). SPECT/CT with yttrium-90 bremsstrahlung photons with
99mTc-MAA in radioembolization with 90Y GATE. Med Phys 37:2943–2950.
microspheres in patients with hepatocellular Rong, X., Du, Y., Frey, E.C. (2012a). A
cancer. J Nucl Med 50:688–692. method for energy window optimiza-
Heard, S., Flux, G.D., Guy, M.J., Ott, R.J. (2004). tion for quantitative tasks that includes
Monte Carlo simulation of 90Y bremsstrah- the effects of model-mismatch on bias:
lung imaging. IEEE Nucl Sci Symp Conf Application to Y-90 bremsstrahlung
Record 6:3579–3583. SPECT imaging. PhysMed Biol 57(12):
Ho, S. et al. (1996). Partition model for estimating 3711–3725.
radiation doses from yttrium-90 microspheres Rong, X. et al. (2012b). Development and
in treating hepatic tumours. Eur J Nucl Med evaluation of an improved quantitative 90Y
23:947–952. bremsstrahlung SPECT method. Med Phys
Ho, S. et al. (1997). Clinical evaluation of the parti- 39:2346–2358.
tion model for estimating radiation doses from Shen, S. et al. (1994a). Planar gamma
yttrium-90 microspheres in the treatment of camera imaging and quantitation of
hepatic cancer. Eur J Nucl Med 24:293–298. yttrium-90 bremsstrahlung. J Nucl Med
Ilhan, H. et al. (2015). Predictive value of 99mTc- 35:1381–1389.
MAA SPECT for 90Y-labeled resin micro- Shen, S., DeNardo, G.L., DeNardo, S.J. (1994b).
sphere distribution in radioembolization of Quantitative bremsstrahlung imaging of
primary and secondary hepatic tumors. J Nucl yttrium-90 using a Wiener filter. Med Phys
Med 56:1654–1660. 21(9):1409–1417.
Kappadath, S.C., Mikell, J., Mourtada, F., Siman, W., Mikell, J., Kappadath, S.C. (2016).
Mahvash, A. (2014). Voxel-based dosimetry Energy window-based scatter compensa-
and radiobiological modeling of HCC tumor tion method to improve image quality
response after 90Y microsphere therapy. and quantification of 90Y bremsstrahlung
J Nucl Med 55:151P. imaging. Med Phys 43:.
Kao, Y.H. et al. (2013). Post-radioembolization Walrand, S. et al. (2014). Optimal design of
yttrium-90 PET/CT—Part 2: Dose–response anger camera for bremsstrahlung imag-
and tumor predictive dosimetry for resin ing: Monte Carlo evaluation. Front
microspheres. EJNMMI Res 3:57. Oncol 4:149.
King, M.A., Coleman, M., Penney, B.C., Glick, S.J. Wondergem, M. et al. (2013). 99mTc-macroaggre-
(1991). Activity quantitation in SPECT: A study gated albumin poorly predicts the intrahe-
of prereconstruction Metz filtering and use patic distribution of 90Y resin microspheres
of the scatter degradation factor. Med Phys in hepatic radioembolization. J Nucl Med
18(2):184–189. 54:1294–1301.
11
Quantitative postradioembolization
imaging using PET/CT

MARCO D’ARIENZO, LUCA FILIPPI, AND ORESTE BAGNI

11.1 Introduction 229 11.5.2 PET absolute activity calibration 237


11.2 Emission of b+ particles via internal 11.5.3 Spatial resolution 237
pair production in the 0+ –0+ transition 11.5.4 Recovery coefficients 238
of 90Zr 230 11.5.5 Degrading factors in 90 Y PET
11.3 Current practice in quantitative imaging 241
postradioembolization quantitative 11.5.6 Image reconstruction 243
imaging with 90 Y 231 11.6 Preparation of a calibrated phantom
11.4 Activity measurements for for 90 Y-PET studies 244
quantitative imaging 233 11.7 Discussion and summary 245
11.4.1 Measurements of 90 Y chloride 233 11.8 Conclusions 245
11.4.2 Measurements of 90 Y microsphere 234 References 247
11.5 Performance characteristics of PET
scanners 235
11.5.1 PET sensitivity 235

2006), where the uncertainties associated with the


11.1 INtrODUCtION beta energies are at k = 1. The average energy of β–
emissions from 90Y is 926.7 (8) keV (Bé et al., 2006),
Over the last decade, the transarterial radioem- with a mean tissue penetration of 2.5 mm and a
bolization with 90Y microspheres has emerged as maximum of 11 mm.
a mainstream treatment modality for inoperable The reader is directed to refer Chapter 1
hepatic malignancies. Radioembolization with for a comprehensive overview of the clinical
90Y is a liver-directed therapy with the potential aspects of radioembolization with 90Y micro-
of delivering a high radiation dose directly to spheres. However, it is worth pointing out
liver tumors in the form of localized β radiation, again that there are presently two clinically
capable of sparing healthy tissue. Due to its long β available 90Y microsphere devices: glass micro-
particle range, 90Y allows for reasonably uniform spheres, sold under the commercial name of
irradiation of large tumors commonly expressing TheraSpheres (TheraSphere®, Nordion Inc. for
heterogeneous perfusion and hypoxia. BTG International, Ottawa, ON, Canada), and
90Y is a pure β-emitter that decays to the ground resin microspheres, sold under the commer-
state of 90Zr with a maximum beta energy of 2279.8 cial name of SIR-Spheres (SIR-Spheres®, Sirtex
(17) keV and a half-life of 2.6684 (13) days (Bé et al., Medical Limited, North Sydney, Australia).

229
230 Quantitative postradioembolization imaging using PET/CT

Although 90Y has been traditionally considered rise to the transfer of radiation energy to an atomic
as a pure β–emitter, the decay of this radionuclide electron in the orbital cloud by internal conversion;
has a minor branch to the 0+ first excited state (2) a transition may occur via electron–positron
of stable 90Zr at 1.76 MeV, which is followed by a internal pair production (if the energy of the pro-
β+/β– emission with an extremely small branching cess is greater than 2me c 2, i.e., 1.022 MeV, where me
ratio. In recent years, a number of authors showed is the mass of the electron); (3) two-photon emis-
that 90Y internal pair production can be imaged by sion, which generally is associated with a negligibly
positron emission tomography (PET), with results small yield. In 1955, Ford first predicted an excited
superior to that of bremsstrahlung single-photon state (0+ state) of 90Zr (Ford, 1955) that was experi-
emission computed tomography (SPECT) for eval- mentally proven by Johnson et al. (1955) in the same
uating the 90Y microsphere biodistribution after period. Using a 90Y source beta-decaying to 90Zr, the
therapeutic radioembolization. The major issue authors discovered a transition at 1.76 MeV followed
associated with 90Y PET imaging is the extremely by a β+/β– emission with an extremely small branch-
small emission probability of the β+ particles. ing ratio (Figure 11.1). These authors also reported
Therefore, in order to obtain acceptable image the probability of pair creation per beta decay as
quality, a high concentration of the 90Y is required. w p / wβ = (2 ± 1) × 10−4.
In liver radioembolization, typical injected activ- One year later, Greenberg and Deutsch (1956)
ity ranges from one to several GBq and the total evaluated the entity of internal pair creation by
amount of radioactivity is concentrated within the assessing the number of positron emissions rela-
liver or even in hepatic segments. Hence, high 90Y tive to the main beta spectrum. From their experi-
concentrations may be obtained with this tech- ment, the positron branching ratio was determined
nique and the PET imaging of 90Y has proven to to be w p / wβ = (3.6 ± 0.9) × 10−5. Later, Langhoff and
be a viable imaging option, possibly leading to the Hennies (1961) determined, with a scintillation
accurate evaluation of in vivo dosimetry. coincidence spectrometer, the positron branch-
Quantitative imaging with 90Y PET/CT is a ing ratio for 90Y to be w p / wβ = (3.4 ± 0.4) × 10−5. In
relatively new imaging strategy and imaging capa- recent years, Selwyn et al. (2007) used a high-purity
bilities are strongly related to PET scanner per- germanium detector to determine the internal pair
formance. Despite the number of phantom and
patient studies that have proven feasibility and
superior quality of quantitative imaging using 2-
0 E = 2,279.800 KeV
PET/CT, to date no standardized imaging protocol 90
39Y51
has been proposed. This chapter gives insight into β-02
-
β01
the major issues related to quantitative postradio- 2+ E = 2,186.282 KeV
embolization imaging techniques using PET/CT. 2
0+ E = 1,760.720 KeV
1
β-00

11.2 EMISSION OF a+ PartICLES


VIa INtErNaL PaIr
PrODUCtION IN tHE 0+–0+ 0
0+ E = 0 Stable
traNSItION OF 90Zr 90
40Zr50

In the past, there has been a great interest in elec- Figure 11.1 Decay scheme of 90Y. 90Y undergoes
tric monopole transitions (E0) in certain nuclei β− decay to 90Zr with a half-life of 2.6684 (13) days
and maximum beta energy of 2279.8 (17) keV.
(e.g., 16O, 40Ca, 72Ge, 90Zr), occurring when there
(From Bé et al., Monographie BIPM-5., 2006.) In
is no angular momentum change between initial addition, 90Y has a minor branch to the 0+ first
and final nuclear states and no parity change. For excited state of stable 90Zr at 1.76 MeV, which is
spin-zero to spin-zero transitions, single gamma followed by a β+/β – emission with an extremely
emission is strictly forbidden and three alterna- small branching ratio, namely (3.186 ± 0.047)∙10 -5.
tive processes may occur: (1) a transition may give (From Selwyn et al., Appl Radiat Isot., 65, 2007.)
11.3 Current practice in postradioembolization quantitative imaging with 90Y 231

Table 11.1 Probability of pair creation per beta decay measured in previous
and recent literature studies
references wp / wβ Detector
Johnson et al. (1955) −4
(2 ± 1) × 10 NaI
Greenberg and Deutsch (1956) (3.6 ± 0.9) × 10−5 NaI
Langhoff and Hennies (1961) (3.4 ± 0.4) × 10−5 NaI
Selwyn et al. (2007) (3.186 ± 0.047) × 10−5 HPGe

production branching ratio of the 0+–0+ transi- authors assessed the 90Y distribution on a Derenzo
tion of 90Zr. The basic measurement technique phantom using a micro-PET scanner equipped
required counting the gross number of gammas with bismuth germanate (BGO) crystals. The study
detected within a 511 keV (annihilation) peak and showed the remarkable resolution and quantita-
subtracting the bremsstrahlung continuum, envi- tive accuracy of positron tomography with 90Y.
ronmental continuum, and environmental peak The first clinical study based on the PET detection
at 511 keV. The authors identified the branching of 90Y internal pair production was carried out by
ratio to be w p / wβ = (3.186 ± 0.047) × 10−5 with a Lhommel et al. (2009). Using a Philips Gemini TF
near 10-fold increase in precision compared with LYSO crystal ToF PET/CT scanner, the authors
previous measurements. A detailed description of demonstrated the feasibility of in vivo imaging of
90Y-labeled resin microspheres in a patient treated
the experiments performed in the past to study the
emission of β+ particles via internal pair produc- with radioembolization for colorectal liver metas-
tion in the 0+–0+ transition of 90Zr can be found tases. With a 30-minute acquisition, the authors
in D’Arienzo (2013). For decades, this transition obtained high-resolution images of the radiophar-
of 90Y was not exploited in nuclear medicine. Only maceutical, clearly surpassing the image quality of
recently, a number of authors have used the small traditional bremsstrahlung SPECT. Of note, the
positronic emission of 90Y to obtain high-resolution authors used a 2.5-mm copper ring to avoid detec-
PET images of 90Y-labeled microspheres. Table 11.1 tor saturation due to bremsstrahlung radiation.
reports the experimental values for the internal In a similar study in 2010, the same authors dem-
pair production branching ratio of the 0+/0+ tran- onstrated the possibility of performing 90Y ToF
sition of 90Zr. PET-based dosimetry in vivo in a patient receiv-
The accuracy of quantitative 90Y PET/CT imag- ing radioembolization with resin microspheres
ing is strongly related to the knowledge of the inter- for metastatic liver disease (Lhommel et al., 2010).
nal pair production branching ratio. It is essential Their pioneering studies led to an explosion in
in the near future that additional measurements of scientific research in this field. Werner et al.
the β+ emission probability will be performed in (2010) performed similar phantom studies using
order to validate the intensity of the branching ratio a non-ToF PET/CT scanner (Biograph Hi-Rez 16,
and possibly reduce the related standard uncer- Siemens Healthcare, Knoxville, TN, USA) showing
tainty (currently about 1.5%; Selwyn et al., 2007). for the first time that 90Y PET/CT scan was possi-
ble even without a ToF. Furthermore, they did not
encounter issues with detector saturation. A simi-
11.3 CUrrENt PraCtICE IN lar study by Wissmeyer et al. (2011) demonstrated
the feasibility of 90Y imaging following liver radio-
POStraDIOEMBOLIZatION
embolization using a Philips Gemini ToF PET/MR
QUaNtItatIVE IMaGING hybrid scanner. Encouraging results with non-ToF
WItH 90Y scanners were obtained by subsequent studies
performed by Gates et al. (2011), with a Siemens
The first phantom study on 90Y PET/CT imaging Biograph 40 with cerium-doped lutetium ortho-
was performed in 2004 by Nickles et al. (2004). The silicate (LSO crystal) detectors, by D’Arienzo et al.
232 Quantitative postradioembolization imaging using PET/CT

(2012) and Bagni et al. (2012) using a GE Discovery body phantom. Each center was asked to perform
ST PET/CT scanner provided with BGO crystals 90Y PET acquisitions over a 7-day period (activ-

and by Fourkal et al. (2013). Other recent studies ity range: 0.5–3.0 GBq). Imaging consisted of two
confirmed the possibilities of perfoming 90Y acqui- overlapping bed positions, each of 15- to 20-minute
sitions with non-ToF PET scanners (Ng et al., 2013; duration. Data were analyzed at a core laboratory
Tapp et al., 2014). Nonetheless, extensive research for consistent processing. Based on these data, the
to date has provided ample evidence that quanti- authors concluded that GE Healthcare and Siemens
tative 90Y PET imaging is likely to improve if ToF ToF systems are suitable for quantitative postradio-
PET/CT scanners are used (van Elmbt et al., 2011; embolization imaging using PET/CT. Quantitative
Willowson et al., 2012, 2015; Carlier et al., 2013, accuracy non-ToF scanners from GE Healthcare
2015; Attarwala et al., 2014; Martí-Climent et al., and Siemens was inferior to ToF systems. Greater
2014). This is explained by the higher sensitivity deviations were obtained on the Philips systems at
and spatial resolution associated with ToF PET/ low count rates (Willowson et al., 2015).
CT systems, resulting in the potential to increase It is worth noting that most of the current
the image signal-to-noise ratio (Lewellen, 1998; generation ToF PET scanners are equipped with
Conti, 2009; Surti, 2014; Surti and Karp, 2016). lutitium-based crystals, e.g., cerium-doped LYSO
Broadly speaking, image generation in non-ToF or cerium-doped LSO. Lutitium-based compounds
3D PET imaging is based on the detection of coin- have desirable properties for ToF PET imaging
cident lines-of-response (LORs) at many angles. due to the high detection efficiency (density rang-
Tomographic images are then generated through ing from 6.7 to 8.3 g/cm3) and excellent temporal
using iterative reconstruction methods. The con- resolution (Conti et al., 2009). The major draw-
cept of ToF is based on the capability of fast elec- back with these scintillators is that the presence of
tronics and scintillators to measure the difference the naturally occurring isotope 176Lu, which gives
in arrival times between two coincident photons, rise to background count rates within the crystal.
thus providing additional information related to While this is not likely to be an issue with tradi-
the location of the decay of a coincidence event. tional 18FDG-PET imaging due to the high positron
While the reconstruction techniques associated abundance, it may hinder accurate quantification
with ToF are outside the scope of this chapter, the under conditions of low counts and high random
end result is improved image quality and quan- fraction, as in 90Y PET/CT imaging. This issue will
tification with shorter scan times in the case of be further discussed in Section 11.5.5.
traditional oncologic imaging with fludeoxyglu- Recent research has shown that quantitative
cose (18F-FDG). For 90Y imaging, due to the low postradioembolization imaging using PET/CT may
branching fraction for positron emission, ToF pave the way for accurate in vivo dosimetry stud-
imaging allows for acceptable image quality and ies. In particular, a number of authors suggested
quantification. that dose–volume histograms (DVH) obtained
The outcome of a recent multicenter compari- from 90Y PET/CT images may be an important pre-
son of quantitative 90Y PET/CT for dosimetric dictor of dose response (D’Arienzo et al., 2013; Kao
evaluation after radioembolization with resin et al., 2013, Fowler et al., 2016). In another study,
microspheres (quantitative uptake evaluation in Ng et al. (2013) extended the analysis of DVHs
SIR-Spheres therapy [QUEST] phantom study, obtained from quantitative data to derive the bio-
Willowson et al., 2015) provided support for the logically effective dose in patients treated with 90Y
hypothesis that ToF PET/CT scanners are capa- radioembolization.
ble of achieving higher accuracy in quantitative Despite encouraging results reported by the
90Y imaging. The QUEST phantom study investi- aforementioned studies, it is worth noting that
gated and compared the quantitative accuracy of quantitative postradioembolization imaging
90Y imaging across different generation ToF and using 90Y PET/CT is subject to inherent limita-
non-ToF PET/CT scanners. Quantitative accuracy tions, mostly attributable to acquisition at low
was assessed by 47 international sites (for a total count rates with a high random fraction. In addi-
of 69 scanners, 37 with ToF mode) following a tion to the presence of background radiation
strict experimental and imaging protocol based due to 176Lu in lutitium-based crystals, several
on acquisitions of the NEMA 2007/IEC 2008 PET authors pointed out a possible reconstruction
11.4 Activity measurements for quantitative imaging / 11.4.1 Measurements of 90Y chloride 233

bias due to the production of negative pixels where a calibrated amount of radiopharmaceutical is
resulting from random-coincidence correction. inserted into the phantom. Therefore, accurate activ-
In fact, commercial software packages truncate ity measurements at a clinical level are the backbone
the negative pixel values before iterative recon- of quantitative imaging as any uncertainties in the
struction, thus leading to a significant posi- initially measured activity concentration will propa-
tive bias in 90Y PET imaging (Tapp et al., 2014; gate to an uncertainty in final clinical quantification.
Walrand et al., 2015). One of the major drawbacks of quantitative
Of additional concern is the lower contrast imaging with 90Y microspheres is related to the
recovery obtained with 90Y PET imaging compared quick microsphere sedimentation over time.
with 18FDG PET, which remains at least partly Therefore, in order to have a homogeneous solu-
unexplained. A deeper insight into this problem is tion, phantom studies dedicated to 90Y PET/CT
provided in Section 11.5.10 (Willowson et al., 2012; imaging are generally performed using 90Y chloride
Carlier et al., 2013, 2015). (90YCl3) instead of 90Y microspheres. In this section,
Another aspect to consider is that many PET the issues related to activity measurements of 90Y at
workstations do not offer 90Y as a viable radionu- a clinical level (both in form of 90Y chloride and 90Y
clide choice for PET scans. In such a case, acqui- microspheres) are described.
sitions can be performed selecting one of the
positron emitters available in the software package 11.4.1 MEASUREMENTS OF 90Y
and accounting for the correct branching ratio and CHLORIDE
half-life (Fourkal et al., 2013; Pasciak et al., 2014a;
Carlier et al., 2015). The instrument typically used to measure the
However, in order to set up a routine acquisi- administered activity to patients in nuclear med-
tion protocol for quantitative postradioemboli- icine procedures is the radionuclide dose calibra-
zation imaging using 90Y PET/CT, preliminary tor. Recent (Fenwick et al., 2014; Ferreira et al.,
phantom studies are required, aimed to character- 2016; Kossert et al., 2016) and previous (Woods
ize and optimize the performance of the scanner et al., 1996) findings reported the difficulties
being used. Sections 11.4–11.6 offer insight into the of measuring 90Y chloride and other beta emit-
procedures needed to setup a clinical protocol for ters using clinically available ionization cham-
quantitative postradioembolization imaging using bers. This is because dose calibrators available
PET/CT. in medicine perform activity measurements of
beta emitting radionuclides indirectly by detect-
ing bremsstrahlung emissions. Bremsstrahlung
production is highly dependent on the source
11.4 aCtIVItY MEaSUrEMENtS material, its container, and the calibrator cham-
ber wall. The ionization current also depends
FOr QUaNtItatIVE
on the probability of electron detection within
IMaGING the chamber, which varies with electron energy
and individual dose calibrator construction.
Accurate and precise activity measurements are an Moreover, slight variations in the container wall
essential prerequisite of therapy with 90Y micro- thickness, solution volume, or location within
spheres. The International Atomic Energy Agency the well can lead to an increase in the overall
(IAEA) Basic Safety Standards states that (IAEA, assay uncertainty when using the manufacturer
1996): “the calibration of sources used for medical supplied calibration factor, which is typically
exposure shall be traceable to a Standard dosim- traceable to national standards.
etry laboratory” and “unsealed sources for nuclear A proper quality control program should
medicine procedures shall be calibrated in terms of be in place for any clinically used radionuclide
activity of the radiopharmaceutical to be adminis- dose calibrator, including a track record of cali-
trated, the activity being determined and recorded brations and consistent daily quality assurance
at the time of administration.” Furthermore, per- measurements. For activity measurements of
formance characteristics of a PET scanner are gen- 90YCl at a clinical level, it is expected that radio-
3
erally assessed through dedicated phantom studies nuclide dose calibrators provide accuracy within
234 Quantitative postradioembolization imaging using PET/CT

±5% (at k = 2 level) (Gadd et al., 2006; AAPM, delivery uncertainty on the order of 20% can be
2012). However, if the activity is determined expected. One of the largest components of the total
by a National Metrology Institute, uncertainty uncertainty is related to the initial activity measure-
on the activity concentration can be reduced ment of the treatment dosage. (Dezarn et al., 2011).
significantly. Primary activity standards for 90Y At a clinical level, activity measurements
are widely available and expanded uncertainties of 90Y are made using radionuclide calibrators
(k = 2 or two standard deviations) of less than 1% traceable to a national standards laboratory
can be achieved (Zimmerman and Ratel, 2005; for the geometry being measured. However,
Dezarn et al., 2011). dose calibrator response to 90Y radioemboliza-
tion is far from being ideal due to the variation
of microsphere distribution in the vial, sample
11.4.2 MEASUREMENTS OF 90Y geometry, and possible variations in the con-
MICROSPHERE tainer wall thickness. Furthermore, a number
of studies showed that in the absence of a well-
Clinical measurement of beta particles emitted by defined local calibration procedure, variations in
90Y microspheres poses additional problems related activity measurements on the order of 10% can
to measurement geometry and homogeneity of the occur (Dezarn and Kennedy, 2007a, 2007b). In
sample (microspheres in solution settle over time, particular, microsphere sedimentation is a major
with measurements affected as spheres settle). issue during the activity measurement. Activity
Currently, there is no traceability to national and measurements performed on a sample of settled
international standards for 90Y microspheres. As a microspheres will likely differ from activity mea-
consequence, there is an urgent need to establish surements performed with the same activity of
a capability for accurately measuring the activ- 90YCl uniformly dispersed in the aqueous solu-
3
ity of 90Y microspheres to traceable measurement tion. The effect of microsphere sedimentation on
standards. the activity measurement is shown in Figure 11.2.
A number of recent studies have been dedicated The dashed curve (indicated with triangles) was
to the standardization of 90Y and determination of obtained using a clinical dose calibrator avail-
calibration factors for 90Y microspheres. The reader able at IFO-Regina Elena Hospital, Rome, Italy.
is referred to Lourenço et al. (2015), Ferreira et al. A vial containing a 5 mL solution of sterile water
(2016), and Thiam et al. (2015). At present, BTG par- uniformly mixed with 3 GBq of 90Y resin micro-
ticipates in the NIST Radioactivity Measurement spheres was measured after shaking the vial
Assurance Program (NRMAP) and NIST main- (microspheres resuspended in the sample). The
tains a secondary measurement standard for the same measurement (dotted curve, indicated with
routine calibration of TheraSphere. SIR-Spheres boxes) was repeated using the NPL-CRC ion-
do not have a NIST traceable calibration, however, ization chamber radionuclide calibrator avail-
activity measurements of 90Y SIR-Spheres have able at the Italian National Institute of Ionizing
been performed at the Australian Nuclear Science Radiation Metrology (INMRI). Finally, the solid
and Technology Organization (ANSTO) and the line (indicated with circles) was obtained after
Australian Radiopharmaceuticals and Industrials the removal of the liquid buffer from the sam-
(ARI) (Dezarn et al., 2011). ple (only microspheres present in the vial, no
At present, each vial of resin microspheres is sedimentation). This provides support for the
calibrated individually within a ±10% range. When hypothesis that a reliable and reproducible mea-
new clinical centers start using 90Y resin micro- surement should be performed after at least a
spheres, the manufacturer provides an activity 200-second waiting time to allow the complete
from the batch report for the first three microsphere sedimentation of microspheres. If the activity
vials shipped. This is needed to allow the clinical measurement is performed at the very begin-
user to normalize the ionization chamber avail- ning of the measurement, it is likely that the total
able on-site to the same calibration as the manu- activity would be underestimated, resulting in
facturer. However, activity measurements of 90Y possible radiopharmaceutical overdose to the
microspheres at a clinical level remain quite critical. patient. This finding was recently confirmed by
Recent literature findings showed that a total dose Ferreira et al. (2016).
11.5 Performance characteristics of PET scanners / 11.5.1 PET sensitivity 235

1.01

1.00

Normalized reading (a.u.)


0.99

0.98

0.97 ≈–6%

Stability after about 200 s


0.96

0.95
5 cc vial, ENEA-INMRI ionization chamber
0.94 2 cc vial, ENEA-INMRI ionization chamber
5 cc vial, hospital dose calibrator
0.93
0 50 100 150 200 250 300 350 400
Time (s)

Figure 11.2 Measurements of 90Y microspheres. The dashed line was obtained using the clinical
dose calibrator (IFO-Regina Elena Hospital, Rome). A vial containing 5 mL solution of sterile water
uniformly mixed with 3 GBq of 90Y-microspheres was measured after shaking the vial (microspheres
resuspended in the sample). The same measurement (dotted line) was repeated using the NPL-CRC
ionization chamber radionuclide calibrator available at ENEA-INMRI. The solid line was obtained after
the removal of liquid buffer from the sample (only microspheres present in the vial, no sedimentation).
ENEA-INMRI, Italian National agency for new technologies, Energy and sustainable economic develop-
ment-National Institute of ionizing Radiation Metrology; IFO, Istituti Fisioterapici Ospitalieri; NPL-CRC,
National Physics Laboratory-Capintec Radionuclide Calibrators.

bequerel. Sensitivity depends on a number of


11.5 PErFOrMaNCE physical and geometrical factors, among which
CHaraCtErIStICS OF PEt are intrinsic efficiency, geometric efficiency, win-
SCaNNErS dow settings, and the dead time of the system. It is
worth noting that detection efficiency depends on
A major objective of PET studies with 90Y PET/ the scintillation decay time, density, atomic num-
CT is to obtain good quality images and a signifi- ber, and thickness of the detector material. The
cant level of detail despite the very low β+ emis- sensitivity of a scanner is highest at the center of
sion probability of 90Y. Reaching this goal depends the axial FOV and gradually decreases toward the
strongly on how well the scanner performs in the periphery (Cherry et al., 2012). Sensitivity mea-
image formation. Several parameters associated surements are generally performed according to
with the scanner may have a significant impact on the NEMA NU 2-2007 procedure (NEMA, 2007).
the image formation. A description of the major Since the emitted positrons annihilate with elec-
parameters possibly influencing quantitative post- trons to create a pair of γ-rays, a sufficient amount
radioembolization imaging using PET/CT is given of material must surround the source to ensure
below. annihilation. However, the surrounding mate-
rial also attenuates the emitted γ-rays. Therefore,
11.5.1 PET SENSITIVITY in order to arrive at an attenuation-free value of
the sensitivity, successive measurements must be
The sensitivity of a PET device is defined as the rate repeated with a uniform line source surrounded
of true coincidence events per unit time detected by a number of known absorbers. The sensitiv-
by the scanner per unit of radioactivity concentra- ity with no absorber is then extrapolated from
tion present in a given source phantom. Sensitivity these measurements. To this purpose, a National
is generally expressed in counts per second, per Electrical Manufacturers Association (NEMA)
236 Quantitative postradioembolization imaging using PET/CT

PET Sensitivity Phantom™ can be used, consisting to the activity of the source. The absolute sensitiv-
of a set of six concentric aluminum tubes. ity for 90Y was 0.403 and 0.388 counts-per-second
PET acquisitions can be performed either in (cps)/MBq with the line source centered on the FOV
two-dimensional (2D) or in three-dimensional (3D) and 10-cm off-center, respectively. Acquisitions were
acquisition mode. In 2D acquisition mode, axial col- performed using a Biograph mCT-TrueV scanner
limation is obtained by positioning tungsten septa with ToF. In another work, Bagni et al. (2012) mea-
between the detector rings. Septa are 1–2 mm thick sured the PET scanner sensitivity to 90Y following
and extend approximately 8–12 cm radially and the same procedure. PET images were acquired with
are generally used to improve the image quality by a GE Discovery ST PET/CT scanner. The NEMA
reducing the detection of scatter and random coin- sensitivity phantom made of six fillable tubes was
cidence events (Peller et al., 2012). However, most used, with the inner tube homogenously filled with
true coincidence events are also reduced. As a con- water mixed with 300 MBq calibrated activity of 90Y.
sequence, the PET scanner sensitivity will decrease The system sensitivity was measured at two radial
when scans are performed in the 2D acquisition positions, at a 0- and 10-cm radial offset from the
mode. To maximize the sensitivity of a PET scanner, center of the transaxial FOV. The measured absolute
septa can be removed with the aim to increase the sensitivity of the scanner for detecting annihilation
number of detected events. This is known as 3D PET photons is 0.409 and 0.577 cps/MBq at 0 and 10 cm
mode. In the 3D mode, the system sensitivity is sig- offset, respectively. In the same study, the authors
nificantly higher than the 2D acquisition mode, with assessed the sensitivity of the PET scanner during
a sensitivity greatest at the axial center of the system. the 2D acquisition mode. The authors found that
Sensitivity measurements for 90Y PET studies can for 2D acquisitions the scanner sensitivity is about
be performed following the NEMA NU 2-2007 pro- one order of magnitude lower than 3D mode (0.076
cedure, as reported by multiple authors. In a recent and 0.077 cps/MBq at 0 and 10 cm radial offset from
work by Martí-Climent et al. (2014), the authors used the center of the transaxial FOV, respectively). The
a polyethylene tube (1 mm internal diameter) filled different impact of 2D and 3D acquisition modes
with a 90Y solution of 879 MBq and inserted into the in 90Y PET imaging is shown in Figure 11.3. The 3D
NEMA PET Sensitivity Phantom. Each acquisition sensitivity values obtained by Bagni et al. (2012) can
lasted 300 s. The system sensitivity was determined be compared with those found by Ng et al. (2013),
by the ratio of true rate events without absorbing 0.32 cps/MBq, and Werner et al. (2010), 0.72 cps/
material, obtained by extrapolation, with respect MBq using a GE Discovery STE PET/CT scanner

Figure 11.3 International Electrotechnical Commission (IEC) body phantom imaged with 2D (left)
and 3D (right) acquisition modes. The image is highly contrasted as it is obtained with a lesion-
to-background ratio of 30:1. With such concentrations, the difference in the signal-to-noise ratio
between the 2D and the 3D mode can be clearly seen, with 2D acquisitions showing dishomogeneity
areas and blurred margins. (From Bagni et al., Nucl Med Commun., 33, 2012. With permission.)
11.5 Performance characteristics of PET scanners / 11.5.3 Spatial resolution 237

and a Siemens Biograph 16 HiRez PET scanner, radionuclide option. Therefore, other radionu-
respectively. clides are generally used for PET absolute activity
Ultimately, it is worth stressing that because of calibration. In order to obtain 90Y activity con-
the very low emission probability of positrons from centration in terms of kBq/cm3, counts need to be
90Y, sensitivity is greatly reduced compared with ultimately rescaled by the ratio of the β+ emission
18F-FDG PET, which is several orders of magnitude probability of the used radionuclide (wβX+) and that
90
greater both in 2D mode (≈1–2 cps/kBq) and in of 90Y (wβ+ Y). This procedure allows a new calibra-
90
3D mode (≈5–15 cps/kBq) (Peller et al., 2012). In tion factor, f = wβ+ Y / wβX+, to be obtained for correct
particular, Werner et al. (2010) found that the sen- 90Y quantification. A number of surrogate radionu-

sitivity for 90Y PET is reduced by a factor of 3.4e-5 clides have been used in published literature (22Na,
in comparison with 18F PET. Similar results were 86 Y, 68Ge, 18F), with 22Na being the most straight-

confirmed by D’Arienzo et al. (2012) who found a forward choice. Of course, an adjusted decay con-
system sensitivity to 90Y (≈0.5 cps/MBq) about four stant must be introduced in order to account for
orders of magnitude lower than that of 18F (≈9 cps/ the different half-life of the selected radionuclide,
kBq). Because of the decreased sensitivity of 90Y and that of 90Y. An extensive description of this
PET compared with 18F, 2D imaging mode is gen- calibration procedure is provided in Pasciak et al.
erally not compatible with 90Y PET imaging. (2014a) along with a list of adjusted decay constants
for selected radionuclides. Of note, a recent study
11.5.2 PET ABSOLUTE ACTIVITY by Fourkal et al. (2013) identified the measurement
CALIBRATION of the calibration factor as being one of the major
sources of uncertainties in the dose measurements
PET absolute activity calibration is also often (together with the uncertainty related to the posi-
referred to as “well counter calibration.” Absolute tive bias due to the intrinsic radioactivity of scan-
activity calibration factors are required to convert ner’s crystals). In the study, the relative standard
pixel values into a measure of absolute activity deviation of the calibration factor was found to be
per voxel. Following the absolute activity calibra- ≈12%. Therefore, it is expected, in the near future,
tion, the voxel intensity in any 90Y PET image is that more accurate measurements of the 90Y β+
divided by the calibration factor to obtain cali- branching ratio will be published.
brated images in terms of kBq/cm3. A standard
source configuration is generally recommended
consisting of a phantom containing a known and 11.5.3 SPATIAL RESOLUTION
homogeneous activity concentration. The latter
can be measured with the on-site dose calibra- The spatial resolution of a system represents its
tor. Traceability to national standards labora- ability to distinguish between two points after
tory for the geometry being measured is essential image reconstruction. The NEMA guideline NU
for activity determination and for uncertainty 2-2007 (NEMA, 2007) describes a standard pro-
reduction. However, if activity is determined by cedure for the measurement of the spatial resolu-
a national laboratory, the final uncertainty can tion. According to the NEMA procedure, spatial
be reduced significantly. Calibration to absolute resolution measurements are performed by imag-
radioactivity concentration is generally accom- ing point sources in air, and then reconstructing
plished by scanning a cylinder or a phantom with images with no smoothing filters (i.e., using a ramp
large volume. The calibration factor, f, is defined filter). Spatial resolution has to be measured in
as (Cherry et al., 2012) the axial slice and in the transverse slice, the lat-
ter both radially and tangentially. Measurements
counts per pixel are performed with a point source consisting of
f=
activity concentration (kBq / cm3 ) a small quantity of concentrated activity inside a
glass capillary with an inside diameter of 1 mm (or
This procedure is well validated for PET imaging less) and an outside diameter of less than 2 mm.
with 18F. However, such a straightforward calibra- Reconstruction should be performed using fil-
tion method is not applicable to 90Y microspheres tered backprojection with no smoothing and pixel
as most scanners do not support 90Y as a viable size should be set below one-third of the expected
238 Quantitative postradioembolization imaging using PET/CT

full width at half maximum (FWHM) in all three ratio of measured activity to true activity in the
dimensions. The spatial resolution in each direc- object, from simple objects of known geometry
tion is then determined in terms of FWHM of (e.g., spheres). Under clinical conditions, the real
one-dimensional response functions of the point activity value can be then obtained by dividing
source. Although spatial resolution measurements the measured activity in the region of interest by
should be performed in nonrealistic clinical con- the RC. RCs are therefore a function of the object
dition (e.g., absence of scatter, attenuation and geometry, size, object-to-background activity
smoothing filters), it provides a best-case com- concentration ratio, and position in field of view.
parison among scanners, indicating the highest RCs are typically assessed using a NEMA IEC
achievable performance. image quality body phantom consisting of a water-
The spatial resolution in 90Y-PET/CT imaging filled cavity with six spherical inserts suspended
has been assessed by a number of authors with dif- by plastic rods of volumes: 0.5, 1.2, 2.6, 5.6, 11.5,
ferent PET scanners. However, it is worth noting and 26.5 mL (inner diameters of 10, 13, 17, 22, 28,
that despite the existence of the NEMA standard and 37 mm). Measurements of RCs in 90Y PET/CT
procedure for the assessment of spatial resolution, imaging have been described by several authors.
it was not used in all literature studies dedicated to As a general rule, partial volume effects were evi-
90Y quantitative imaging. dent in all but the largest NEMA sphere (the lower
Werner et al. (2010) found a resolution of 5.2 ± the object size, the lower the RC). All 90Y PET
0.6 mm (336 × 336 matrix, 8 iterations, 16 subsets) studies show that RCs obtained with 90Y PET/CT
and 7.8 ± 0.5 mm (128 × 128 matrix, 4 iterations, imaging are poorer than those obtained with 18F
8 subsets) using a non-ToF Siemens Biograph PET PET/CT imaging (Werner et al., 2010; D’Arienzo
scanner with LSO detector elements. In a recent et al., 2012; Willowson et al., 2012). Furthermore,
study by Martí-Climent et al. (2014), the spatial ToF-PET scanners are likely to improve contrast
resolution under a number of conditions using a of hot spheres and increase RCs (Willowson et al.,
Siemens Biograph mCT-TrueV ToF scanner with 2012). The recent QUEST study (Willowson et al.,
LSO crystals was measured. The authors obtained 2015) aimed to investigate and compare the quan-
spatial resolution values in the range 2.2–12.1 mm. titative accuracy of 90Y imaging across different
In another study, D’Arienzo et al. (2012) per- PET/CT scanners. The results of the study clearly
formed spatial resolution measurements using a confirmed that partial volume effects dominate
non-ToF BGO PET. With an acquisition matrix of spheres of diameter <20 mm when 90Y PET quan-
256 × 256, at a 1 cm radius, 90Y PET transverse titative imaging is performed with current genera-
and axial spatial resolutions were found to be 5.8 tion ToF scanners from GE, Philips, and Siemens
± 0.9 and 5.0 ± 0.6 mm, respectively. When the (Figure 11.4). For spheres >20 mm in diameter,
source was placed at a 10 cm radius, transverse activity concentrations were consistently underes-
radial, transverse tangential, and axial resolutions timated by about 20%. Of note, non-ToF scanners
were found to be 5.5 ± 0.9, 5.7 ± 0.9, and 7.3 ± from GE Healthcare and Siemens were capable of
1.0 mm, respectively. Similar values were obtained producing accurate measures, but with inferior
by Kao et al. (2013) using a LYSO GE Discovery quantitative recovery compared with ToF systems.
690 (10 mm) and by van Elmbt et al. (2011) using Recovery of activity concentration measured in
different PET scanners, i.e., Philips Gemini with the hot spheres on day 0 of imaging is shown in
GSO crystals (10 mm), Philips Gemini TF with Figure 11.5. In particular for a 37-mm-diameter
LYSO crystals (9.3 mm), and Siemens Ecat Exact object average underestimates of −34% and −27%
HR with BGO detector (10.6 mm). A summary of were found for GE Healthcare and Siemens scan-
the spatial resolution values obtained in published ners, respectively (Willowson et al., 2015).
literature is given in Table 11.2. In the presence of hot background, image qual-
ity can be assessed using the hot contrast recov-
11.5.4 RECOVERY COEFFICIENTS ery coefficient (CRChot), as reported by the NEMA
guidelines (Daube-Witherspoon et al., 2002):
The use of recovery coefficients (RCs) is a simple
and widely used tool for the correction of par- CRC hot =
(Chot − Cbkgd ) − 1
tial volume effects (PVE). RCs are defined as the (ahot − abkgd ) − 1
11.5 Performance characteristics of PET scanners / 11.5.4 Recovery coefficients 239

Table 11.2 Image reconstruction parameters and spatial resolution values in quantitative
postradioembolization imaging studies using 90Y PET/CT

Y
90 Scanner Detector acquisition
reference microspheres manufacturer crystal mode reconstruction resolution
Lhommel et al. Resin Gemini Philips LYSO ToF 2 iterations, 33 –
(2010) microspheres substeps
Werner Resin Biograph LSO Non ToF 8 iterations, 16 6.4 mm
et al. (2010) microspheres Hi-Rez 16 subsets and 4
Siemens iterations, 8
subsets
Gates et al. Glass Biograph 40 LSO Non-ToF 3 iteration, 21 2.5–4 mm
(2011) microspheres Siemens subsets
Wissmeyer Glass Philips Gemini LYSO ToF 3 iterations, 33 –
et al. (2011) microspheres PET/MR subsets
Bagni et al. Resin Discovery ST BGO Non-ToF 2 iterations, 15 6.3 mm
(2011) microspheres GE subsets
Carlier et al. Resin and Biograph mCT LSO ToF and 1 or 3 –
(2013) glass 40 Siemens Non-ToF iterations, 21
microspheres or 24 subsets
Elschot et al. Resin Biograph mCT LSO ToF 3 iterations, 21 –
(2013) microspheres Siemens or 24 subsets
Kao et al. Resin Biograph WO LSO Non-ToF 2 iterations, –
(2012) microspheres Siemens 8 subsets
Kao et al. Resin Discovery 690 LYSO ToF 3 iterations, 10–12 mm
(2013) microspheres GE 18 subsets
van Elmbt Resin Philips Gemini LYSO ToF 3 iterations, 9.3 mm
et al. (2011) microspheres TF 8 substeps
van Elmbt Resin Philips Gemini GSO Non-ToF 3 iterations, 10 mm
et al. (2011) microspheres Power16 8 substeps
van Elmbt Resin Siemens Ecat BGO Non-ToF 3 iterations, 10.6 mm
et al. (2011) microspheres Exact HRb 8 substeps
Martí-Climent Resin Biograph LSO ToF 1–3 iterations, 2.2–12.1
et al. (2014) microspheres mCT-TrueV 21–24 mm
substeps

Figure 11.4 Acquisitions of the NEMA phantom used in the QUEST phantom study (Willowson et al.,
2015) by Oxford University Hospitals NHS Foundation Trust. Images were acquired (from left to right)
at days 0, 3, 5, 7 using a GE Discovery 710 ToF system. Total phantom activity at D0 was about 4.5
GBq. Reconstruction was performed with Q. Clear reconstruction algorithm (beta 4000), matrix size
246 × 256. (Courtesy of Lisa Rowley, Oxford University Hospitals NHS Foundation Trust.)
240 Quantitative postradioembolization imaging using PET/CT

(a) (b)
100 100

80 80

Recovery (%)
60 60
Recovery (%)

40 40
2D OSEM (DSTE)
All pass RR+ToF BGO All Pass
Gauss RR+ToF BGO Gauss
All pass ToF BGO RR
20 Gauss ToF 20 LYSO All Pass
Gauss LYSO Gauss
18F Recovery 18F Recovery

0 0
5 10 15 20 25 30 35 40 5 10 15 20 25 30 35 40
Sphere diameter (mm) Sphere diameter (mm)
(c) (d)
100 100

80 80
Recovery (%)

60 60
Recovery (%)

40 40

4i8s BLOB OS TF 3D RAMLA


20 3/4i33s BLOB OS TF 20 18F Recovery
18F Recovery

0 0
5 10 15 20 25 30 35 40 5 10 15 20 25 30 35 40
Sphere diameter (mm) Sphere diameter (mm)
(e) (f )
100 100

80 80

60 60
Recovery (%)
Recovery (%)

40 1i21s All Pass RR+ToF 40


1i21s Gauss RR+ToF
2i21s All Pass RR+ToF
2i21s Gauss RR+ToF
20 3i21s All Pass RR+ToF 20 2D OSEM (normalized)
3i21s Gauss RR+ToF 3D RR (–RAN)
18F Recovery 3D RR (+RAN)
18F Recovery
0 0
5 10 15 20 25 30 35 40 5 10 15 20 25 30 35 40
Sphere diameter (mm) Sphere diameter (mm)

Figure 11.5 Recovery of activity concentration measured in the hot spheres from the QUEST phantom
study. Lines of best fit (y = a + bx) for recovered concentrations in the largest hot sphere at different
concentrations for (a) GE Healthcare ToF systems, (b) GE Healthcare non-ToF systems, (c) Philips ToF
systems, (d) Philips non-ToF systems, (e) Siemens ToF systems, (f) Siemens non-ToF systems (where
+RAN and -RAN correspond to data acquired in “PROMTS + RANDOMS” and “NETTRUES” mode,
respectively, and where -RAN was normalized for analysis). Figure reproduced from Willowson et al.
(2015) under the terms of the Creative Common Attribution 4.0 International License (https://creative-
commons.org/licenses/by/4.0/).
11.5 Performance characteristics of PET scanners / 11.5.5 Degrading factors in 90Y PET 241

where Chot and C bkgd are the average of the counts The major issues in quantitative postradioembo-
measured in the hot sphere region of interest (ROI) lization imaging using PET/CT are related to low
and the average of the counts in all background counting statistics and a high random fraction.
ROIs, respectively, ahot is the true activity concen- These effects are due to a number of factors attrib-
tration in the hot sphere, and abkgd is the true activ- utable both to the physics of the decay and to the
ity concentration in the background. ROIs with detection crystal of a PET scanner.
diameters equal to the physical inner diameters Regarding the limitations related to the phys-
of the spheres must be drawn on the spheres and ics of the decay, 90Y PET images are inherently
throughout the background. A number of authors noisy due to the extremely small positron emis-
have assessed CRChot to quantify the image qual- sion branching fraction of 90Y, resulting in low
ity in 90Y PET acquisitions. Willowson et al. (2012) true coincidence count rate. This is especially true
found that ToF acquisition mode improves the in nontarget anatomical regions, where the 90Y
recovered contrast of hot spheres, an effect which activity concentration will be much lower than in
increases with decreasing sphere diameter. Van treated liver tissue. As a consequence, longer scan
Elmbt et al. (2011) corroborated these findings times than a traditional positron-emitting radio-
pointing out that ToF led to a greater improvement isotope are generally required to obtain satisfactory
in hot contrast for smaller diameters. Furthermore, quality images (by comparison, 18F has a branch-
if ToF reconstructions are used, the hot contrast ing fraction of 967 per 1000 decays). As an exam-
recovery coefficient remains almost constant even ple, acquisition times in 18FDG PET/CT imaging
for shorter acquisitions. are 2–5 minutes per bed position depending on
Finally, it is worth noting that contrast recov- the amount of injected radioactivity, body mass
ery obtained with 90Y imaging is lower than those index, and scanner sensitivity. While longer scans
obtained with 18F. Van Elmbt et al. (2011) analyzed improve image quality, acquisition times for 90Y
PET/CT acquisitions performed with phantoms PET patient studies can’t be increased arbitrarily
filled both with 90Y and 18F. He found that despite and a trade-off between patient comfort and image
scatter fractions being approximately the same, con- quality is required. Current literature studies report
trast recovery with 18F is superior to 90Y. This is partly acquisition times in the range from 10 minutes per
still unexplained. However, some explanations were bed position using a ToF scanner (Tapp et al., 2014)
proposed among which was (1) the effect of high to 40 minutes per bed position with non-ToF scan-
image noise on ordered-subset expectation maxi- ner (Werner et al., 2010). The imaging protocol pro-
mization (OSEM) image reconstruction algorithm. posed in the recent QUEST study consisted of two
Negative pixel values are truncated before itera- overlapping bed positions (to mitigate the triangu-
tive reconstruction leading to a significant positive lar axial sensitivity profile of the scanner) each of
bias in 90Y PET imaging. Another explanation was 15–20 minutes duration, in 3D mode.
(2) possible coincidences arising from pair produc- The high-energy primary β− particle emis-
tion in the scanner crystals by the bremsstrahlung sion of 90Y decay generates a continuous brems-
x-rays above 1.022 MeV. Due to the large atomic strahlung radiation spectrum (Stabin et al., 1994)
numbers of the crystals, the probability of pair pro- that has the potential to degrade both the image
duction in the PET crystals is high (Van Elmbt et quality and the quantitative accuracy of 90Y PET
al., 2011). These pair-production events may have an imaging (maximum energy of β- emissions and,
impact on random corrections. However, this issue therefore, bremsstrahlung photons from 90Y is
has not yet been fully addressed and needs to be fur- 2.28 MeV). In particular, the large flux of brems-
ther investigated in the future. strahlung photons results in a singles count rate
largely exceeding the true coincidence count rate.
The highest bremsstrahlung yield is at energies
11.5.5 DEGRADING FACTORS below 20 keV, which is significantly attenuated
IN 90Y PET QUANTITATIVE by the patient. However, a significant portion of
IMAGING higher energy bremsstrahlung photons are emit-
ted within the acceptance window of PET scanners
It is well known that PET imaging of 90Y provides and has the potential to saturate the PET detectors.
poor quality images compared with 18F imaging. In a previous study, Lhommel et al. (2009) used a
242 Quantitative postradioembolization imaging using PET/CT

homemade 2.5-mm-thick copper ring to reduce order to assess the impact of natural 176Lu radioac-
bremsstrahlung radiation emerging from a patient tivity on the image quality, long acquisition with
administered with 1.3 GBq of 90Y. Of note, typi- no radioactivity present in the field of view is per-
cal administered activities may be in the order of formed to determine the 176Lu background count
several GBq, therefore, possible detector saturation rate.
may not be ignored a priori. One last confounding factor in 90Y PET quanti-
Other degrading factors that significantly con- tative imaging is attributable to scatter correction.
tribute randomly are (1) the detection of random At very low counts, PET images are very noisy
coincidences from two bremsstrahlung photons and the resulting scatter correction might lead
emitted simultaneously; (2) detection of coin- to heavy under- or overestimation of the scatter
cidences from an annihilation 511 keV photon contribution.
and a bremsstrahlung photon emitted simultane- To conclude, bremsstrahlung photons and
ously; and (3) the presence of the naturally occur- prompt gammas are likely to result in a very high
ring isotope 176Lu within the detection crystals random fraction in imaging 90Y on the order of
(i.e., cerium-doped LYSO or cerium-doped LSO) 80% (Willowson et al., 2015) or even higher (Carlier
of PET imaging systems that may generate back- et al., 2015) compared with a typical FDG scan of
ground count rates. There is ample evidence in the 30%–40%. The combination of high random frac-
literature that bremsstrahlung radiation together tion, extremely low true coincidences, and prob-
with the LSO background radiation greatly lematic scatter modeling for low counting statistics
increases the random fraction in quantitative 90Y results in very noisy true coincidence sinograms.
PET imaging. In addition, a well-known problem in PET imag-
Lutetium (Lu)-based scintillators such as LSO ing is the introduction of a positive bias after cor-
and LYSO are widely used in current generation rection for random events (Ahn and Fessler, 2004;
PET detectors (especially in ToF scanners) due to Rahmim et al., 2005; Li and Leahy, 2006). The most
their relatively high stopping power for 511 keV common method of correcting for random coinci-
gamma rays, high light yield, and short decay dences is the real-time or offline subtraction of a
time. However, 2.6% of naturally occurring Lu is delayed coincidence time window from the prompt
the isotope 176Lu (T1/2 ∼ 3.6 × 1010 year), a long-lived signal. In a scenario with low true coincidences
radioactive element undergoing beta decay (maxi- and high random fraction (as in 90Y PET imaging),
mum energy 596 keV) and three major simultane- negative sinogram ray-sum values can be pro-
ous gamma decays at energies 88 keV (15%), 202 duced. These negative sinogram values are often
keV (78%), and 307 keV (94%) (Browne and Junde, truncated in commercial software packages before
1998). While the presence of 176Lu is generally not iterative reconstruction (i.e., become zeroed) thus
an issue with traditional PET radionuclides due to introducing a positive bias. This bias does not have
the high true coincidence count rate (e.g., 18F), this a significant impact for clinical imaging with 18F,
phenomenon is likely to introduce nonnegligible but may become important in 90Y PET imaging.
random events during 90Y PET acquisition, thereby This bias was observed by several authors (Tapp
affecting system performance. et al., 2014; Carlier et al., 2015) and it is possibly
In a PET detector, the β particles emitted from responsible for hot contrast recovery obtained with
Lu-based crystals, given the short range, deliver 90Y being inferior to that obtained with 18F.

most of their energy in the same crystal. On the It is worth mentioning that presently a number
other hand, γ-rays can be detected not only in the of literature studies provided ample evidence that
same crystal where they are generated but also in neither detector saturation (D’Arienzo et al., 2012;
other detector elements. Therefore, the background Bagni et al., 2012; Carlier et al., 2013) nor intrinsic
radiation generated by the radionuclide 176Lu can natural 176Lu (Carlier et al., 2013) radioactivity rep-
contribute to the amount of random and true coin- resents a major issue in 90Y PET quantification at
cidences. The most likely event is the detection of activity concentrations commonly encountered in
coincidences originating from the detection of the liver radioembolization. In particular, the presence
β− in the crystal in which the 176Lu decay occurred of natural 176Lu radioactivity produces a measurable
and one of the prompt γ-rays in another detector but not limiting contribution. Carlier et al. (2013)
crystal (Goertzen et al., 2009). As a general rule, in suggested that emissions from the radionuclide
11.5 Performance characteristics of PET scanners / 11.5.6 Image reconstruction 243

Lu may significantly contribute to random coin-


176 The impact of image reconstruction on 90Y
cidences when 90Y radioactivity concentration is PET quantification has been widely investigated
below 1 MBq mL−1 in the presence of high tumor to by a number of literature studies with varying
background activity concentration ratios. reported success. The general consensus is that
An extensive study on the limitations and the best reconstruction technique will depend on
the accuracy achievable under conditions of low the scanner and the acquisition modality (ToF
counts and high random fraction can be found in or non-ToF). Both Willowson et al. (2012) and
Carlier et al. (2015). Carlier et al. (2013) found that one iteration pro-
vided the most accurate quantification on a ToF
11.5.6 IMAGE RECONSTRUCTION Siemens BioGraph mCT. On the other hand Bagni
et al. (2012) and D’Arienzo et al. (2012) performed
Iterative reconstruction has become the standard acquisitions on a BGO GE discovery ST scanner
for routine clinical PET imaging. However, itera- using two and three iterations, respectively. In
tive algorithms are resource intensive, especially another study van Elmbt et al. (2011) used three
for ToF data, and OSEM algorithms are, therefore, iterations (eight substeps + Gaussian filter) both
commonly used to accelerate reconstruction. As a on a Philips Gemini Power 16 and a Siemens
general rule, the image noise in the reconstructed Ecat Exact HR, while Lhommel et al. (2009) and
images increases as the number of iterations pro- Werner et al. (2010) used two iterations (33 sub-
ceeds (Figure 11.6). On the other hand, image steps, ToF, RR) and eight iterations (16 substeps)
quality is also degraded when OSEM is used with on a Philips Gemini TF and a Siemens BioGraph
a large number of subsets. As a consequence, there Hi-Rez 16, respectively. A summary of the image
is a tradeoff between the number of iterations/sub- reconstruction parameters obtained in published
sets and reconstructed image quality. literature is given in Table 11.2.

1 iteration, 10 substeps 2 iterations, 10 substeps 3 iterations, 10 substeps

4 iteration, 10 substeps 5 iterations, 10 substeps 6 iterations, 10 substeps

Figure 11.6 Effect of number of iterations on the NEMA phantom with six fillable spherical inserts.
Image noise in the reconstructed images increases as the number of iterations proceeds. Images were
obtained using a BGO GE Discovery ST PET scanner.
244 Quantitative postradioembolization imaging using PET/CT

The major outcome of the QUEST phantom should always be prepared using laboratory grade
study is that 90Y imaging capability appears to be chemicals and purified water. Therefore, it is
optimal for Siemens systems using two iterations important to ensure that a favorable chemistry
and 21 subsets with ToF and resolution recovery is used throughout the calibration procedure in
(RR) and an all-pass filter. For GE Healthcare order to have a uniform and stable solution. For
systems, the use of an all-pass filter in conjunc- 90Y PET studies, 90YCl in an aqueous solution of
3
tion with RR and ToF (2 iterations and 24 sub- 0.1 mol dm-3 hydrochloric acid also containing
steps) provided the best quantification results. inactive yttrium at a concentration of about 50 µg
Regarding non-ToF generation scanners, mea- g-1 can be used as a carrier solution. Alternatively,
sures of background provided average devia- diethylenetriaminepentaacetic acid (DTPA) or
tions to within 1%, 5%, and 2% for GE Healthcare ethylenediaminetetraacetic acid (EDTA) at a con-
(all-pass filter, RR + ToF), Philips (4i8s ToF), and centration of about 50 µg∙g-1 can be used to pre-
Siemens (2i21s all-pass filter, RR + ToF) ToF sys- vent radioactive 90Y from sticking to the phantom
tems, respectively. walls and to guarantee a homogeneous radionu-
clide solution.
It is recommended that all containers be pre-
filled with carrier 12 hours prior to addition of
11.6 PrEParatION OF a
radioactive 90YCl3. This will help to “seal” the sur-
CaLIBratED PHaNtOM
face and reduce sticking or plating of activity. All
FOr 90Y-PEt StUDIES containers should be emptied, dried, and the used
carrier discarded before activity is added.
As previously mentioned, most quantitative imag- Activity concentration should be determined
ing studies make use of phantoms and require using a radionuclide dose calibrator traceable to
very accurate knowledge of both the activity and a national standards laboratory for the geom-
the volume of liquid solution inserted into the etry being measured, as previously discussed.
phantoms. The general purpose of quantitative Uncertainty for a typical well-calibrated field
imaging studies with calibrated phantoms is (1) instrument can be expected to be in the region of
to assess uncertainties in the imaging process; ~5% for low-energy gamma emitters (<100 keV)
(2) to quantify accuracy of correction factors and and pure beta emitters, such as 90Y. It is worth
reconstruction algorithms; and (3) to evaluate noting that if activity is determined by a national
scanner performance over time. A recent study laboratory then this uncertainty can be reduced
(Sunderland et al., 2015) demonstrated that tech- significantly.
nical error in phantom filling is one of the pri- As a general rule, preparation of a stock solu-
mary reasons for exclusion of PET/CT scanners tion is recommended. Radioactive 90Y provided
from clinical trials. Section 11.4 was dedicated to by a supplier should be diluted using the carrier
the importance of accurate 90Y activity measure- solution to desired volume and concentration.
ments. Here, we describe further issues that need Activity concentration should be determined
to be addressed for an optimal phantom prepara- by measuring an aliquot of the stock solution in
tion dedicated to quantitative analysis with 90Y terms of activity per unit mass (or volume). This
PET. can then be used to determine the activity of all
Adsorption of radionuclides on the inner walls subsequent sources produced from this stock
of plastic phantoms may lead to inhomogeneous solution.
radionuclide distribution that can negatively Filling of the phantoms should be performed
affect quantitative imaging studies (Park et al., using a calibrated (preferably four decimal places)
2008). Therefore, the preparation of a carrier analytic scientific scale and with routine double or
solution is recommended. The use of tap water triple weighing of the sources. The overall uncer-
should be avoided as minerals and other chemical tainty in the activity concentration determined
impurities might stick to the phantom walls or using this method is dependent on the precision
combine with the radiopharmaceutical changing of the scale being used as well as the accuracy
the radionuclide distribution. Carrier solutions of the method used to determine the activity
11.8 Conclusions 245

concentration of the solution. Radioactivity should of activity concentration in hot spheres is inferior
be dispensed using calibrated pipette devices or to that obtained with 18F, probably due to effects
syringes and ensuring that no air bubbles remain of image noise on the OSEM reconstruction algo-
in the phantom. Either way, it is suggested to flush rithms, which present an inherent nonnegativity
the needle to remove all activity from the syringe constraint. While the intrinsic radioactivity pres-
or the pipette. Again the uncertainty is dependent ent in lutetium-based crystals is a potential limita-
on the precision of the volume measurement as tion, it has been shown to have a negligible impact
well as the activity determined. at the high activity concentrations typically found
If large background volumes are used for cali- in 90Y radioembolization.
bration purposes, the phantom can be filled with Regarding the acquisition time, a 40-minute
nonradioactive water to measure the fillable vol- acquisition is recommended in a clinical scenario,
ume (and to confirm the phantom is watertight acquired as two bed positions, 20 minutes each.
with no leaks). When filling large phantom vol- Figure 11.7 shows different acquisitions performed
umes, a funnel should be used. When the phan- with a GE Discovery ST BGO scanner 2 hours after
tom is nearly full, the funnel can be removed and a the microsphere administration (single bed acqui-
syringe used to complete the filling process thereby sition, 20 minutes). An excellent match between
preventing the spillage of radioactive water from microsphere accumulation and tumor areas was
the background compartment. observed.
Last, the accuracy of quantitative 90Y PET/CT
imaging is strongly related to the measurement
precision of the internal pair production branch-
ing ratio. At present, the β+ emission probabil-
11.7 DISCUSSION aND ity of 90Y is known with an uncertainty around
SUMMarY 1.5% ((3.186 ± 0.047) × 10−5; Selwyn et al., 2007). A
reduction of such uncertainty will lead to a reduc-
PET imaging of 90Y microspheres is a rapidly evolv- tion of the overall uncertainty in the activity quan-
ing field whose features and attributes have not yet tification. This is especially important in light of
been fully addressed. At present, the general con- the current uncertainty in the clinical measure-
sensus is that accurate quantification is possible on ment of 90Y activity using a dose calibrator. In the
a variety of PET scanner models, with or without near future, new experimental measurements of
ToF, although ToF is likely to improve the accu- the internal pair production branching ratio are
racy at lower activity concentrations. Quantitative desirable in order to achieve more accurate quan-
accuracy has been investigated by a number of tification in postradioembolization imaging using
authors using different phantoms with varying PET/CT.
reported success. Regardless of the scanner used,
partial volume effects play a major role in activity
quantification showing a steady decline for spheres
with diameter below 37 mm. It is expected that 11.8 CONCLUSIONS
quantification in small hot regions may be under-
estimated with all current generation scanners to a Currently, a number of clinics have developed their
consistent degree of 15%–20% for a 37-mm-diam- own quantitative imaging procedures using 90Y PET/
eter object (Willowson et al., 2015). Higher quanti- CT. Very often there is a lack of harmonization of
fication accuracy can be achieved for large regions these procedures, most likely because quantitative
uniformly filled with 90Y. Non-ToF generation GE imaging with 90Y PET/CT is a relatively new imaging
Healthcare and Siemens scanners are capable of strategy and further because imaging capabilities
recovering activity concentrations on the order of are strongly related to PET scanner performance.
300 kBq/mL within 2% and 9% of true values, while Ultimately, it is worth stressing that scanner perfor-
the deviation for ToF GE Healthcare and Siemens mance is optimized for 18F imaging and dedicated
scanners can be expected to be in the range 1%–5% imaging protocols are required for accurate 90Y
(Willowson et al., 2015). Furthermore, the recovery PET/CT studies. Careful choice of reconstruction
246 Quantitative postradioembolization imaging using PET/CT

(a)

(b)

(c)

Figure 11.7 Clinical examples of postradioembolization imaging using 90Y-PET showing an excel-
lent match between microsphere accumulation on hepatic lesions. (Left) 90Y-PET fused with CT data.
(Central) CT of the liver parenchyma clearly showing tumor regions. (Right) 90Y-PET. Figure parts (a)
through (c) show different axial levels of the same patient.

parameters has the potential to increase the quan- Comprehensive guidance has yet to be pre-
titative accuracy. However, the final outcome will sented in this field and there is no doubt that an
depend on the scanner and reconstruction software. internationally endorsed protocol on the 90Y PET/
References 247

CT quantitative imaging would lead to further D’Arienzo, M. et al. (2012). 90Y PET-based dosim-
advances in this area. etry after selective internal radiotherapy
treatments. Nucl Med Commun 33:633–640.
REFERENCES D’Arienzo, M. et al. (2013). Absorbed dose to
lesion and clinical outcome after liver radio-
AAPM Task Group 181. (2012). The selection, use, embolization with 90Y microspheres: A case
calibration, and quality assurance of radio- report of PET-based dosimetry. Ann Nucl
nuclide calibrators used in nuclear medicine Med 27:676–680.
report of AAPM Task Group 181. College Daube-Witherspoon, M.E. et al. (2002). PET
Park, MD: American Association of Physicists performance measures using the NEMA NU
in Medicine. 2-2001 standard. J Nucl Med 43:1398–1409.
Ahn, S., Fessler, J.A. (2004). Emission image Dezarn, W. et al. (2011). Recommendations of
reconstruction for randoms-precorrected PET the American Association of Physicists in
allowing negative sinogram values. IEEE Trans Medicine on dosimetry, imaging, and quality
Med Imaging 23:591–601. assurance procedures for 90Y microsphere
Attarwala, A. et al. (2014). Quantitative and brachytherapy in the treatment of hepatic
qualitative assessment of yttrium-90 PET/CT malignancies. Med Phys 38:4824–4845.
imaging. PLoS One 9:e110401. Dezarn, W., Kennedy, A. (2007a). SU-FF-T-380:
Bagni, O. et al. (2012). 90Y-PET for the assess- Significant differences exist across institu-
ment of microsphere biodistribution after tions in 90Y activities compared to reference
selective internal radiotherapy. Nucl Med standard. Med Phys 34:2489.
Commun 33:198–204. Dezarn, W., Kennedy, A. (2007b). Resin 90Y
Bé, M. et al. (2006). Table of radionuclides. microsphere activity measurements for liver
Monographie BIPM-5. Sèvres, Paris: Bureau brachytherapy. Med Phys 34:1896–1900.
International des Poids et Mesures, ISBN Elschot, M. et al. (2013). Quantitative compari-
92-822-2218-7. son of PET and bremsstrahlung SPECT for
Browne, E., Junde H. (1998). Nuclear data imaging the in vivo yttrium-90 microsphere
sheets for A = 176. Nucl Data Sheets distribution after liver radioembolization.
1998;84:337–486. PLoS One 8:e55742.
Carlier, T. et al. (2013). Assessment of acquisition Fenwick, A., Baker, M., Ferreira, K., Keightley, J.
protocols for routine imaging of Y-90 using (2014). Comparison of Y-90 measurements
PET/CT. EJNMMI Res 3:11. in UK hospitals. NPL Report IR 20. United
Carlier, T., Willowson, K., Fourkal, E., Bailey, D., Kingdom: National Physics Laboratory.
Doss, M., Conti, M. (2015). 90Y-PET imaging: Ferreira, K., Fenwick, A., Arinc, A., Johansson,
Exploring limitations and accuracy under L. (2016). Standardisation of 90Y and deter-
conditions of low counts and high random mination of calibration factors for 90Y
fraction. Med Phys 42:4295–4309. microspheres (resin) for the NPL secondary
Cherry, S., Sorenson, J., Phelps, M. (2012). ionisation chamber and a Capintec CRC-25R.
Physics in Nuclear Medicine. Philadelphia, PA: Appl Radiat Isotopes 109:226–230.
Elsevier/Saunders. Ford, K. (1955). Predicted 0+ level of Zr90. Phys
Conti, M. (2009). State of the art and challenges Rev 98:1516–1517.
of time-of-flight PET. Phys Med 25:1–11. Fourkal, E. et al. (2013). 3D inpatient dose
Conti, M., Eriksson, L., Rothfuss, H., Melcher, reconstruction from the PET-CT imaging
C. (2009). Comparison of fast scintillators of 90Y microspheres for metastatic cancer
with TOF PET potential. IEEE Trans Nucl Sci to the liver: Feasibility study. Med Phys
56:926–933. 40:081702.
D’Arienzo, M. (2013). Emission of β+ particles via Fowler, K. et al. (2016). PET/MRI of hepatic 90Y
internal pair production in the 0+–0+ transi- microsphere deposition determines indi-
tion of 90Zr: Historical background and current vidual tumor response. Cardiovasc Intervent
applications. Nucl Med Imaging Atoms 1:2–12. Radiol 39:855–864.
248 Quantitative postradioembolization imaging using PET/CT

Gadd, R. et al. (2006). Protocol for establishing Li, Q., Leahy, R.M. (2006). Statistical model-
and maintaining the calibration of medical ing and reconstruction of randoms precor-
radionuclide calibrators and their quality rected PET data. IEEE Trans Med Imaging
control. Measurement Good Practice Guide 25:1565–1572.
No. 93. Teddington, Middlesex, UK: National Lourenço, V. et al. (2015). Primary standardization
Physical Laboratory. of SIR-Spheres based on the dissolution of the
Gates, V. et al. (2010). Internal pair production 90Y-labeled resin microspheres. Appl Radiat
of 90Y permits hepatic localization of micro- Isot 97:170–176.
spheres using routine PET: Proof of concept. Martí-Climent, J. et al. (2014). PET optimiza-
J Nucl Med 52:72–76. tion for improved assessment and accurate
Goertzen, A., Stout, D., Thompson, C. (2009). A quantification of 90Y-microsphere biodistri-
method for measuring the energy spectrum bution after radioembolization. Med Phys
of coincidence events in positron emission 41:092503.
tomography. Phys Med Biol 55:535–549. Mo, L. et al. (2005). Development of activity
Greenberg, J.S., Deutsch, M. (1956). Positrons standard for 90Y microspheres. Appl Radiat
from the decay of P32 and Y90. Phys Rev Isot 63:193–199.
102:415–421. National Electrical Manufacturers Association.
IAEA. (1996). International basic safety standards (2007). NEMA NU 2-2007 Performance
for protection against ionizing radiation and for Measurements of Positron Emission
the safety of radiation sources. International Tomographs Arlington, VA: NEMA.
Atomic Energy Agency, Vienna, Austria. Ng, S.C. et al. (2013). Patient dosimetry for 90Y
Johnson, O., Johnson, R., Langer, L. (1955). selective internal radiation treatment based
Evidence for a 0+ first excited state in Zr90. on 90Y PET imaging. J Appl Clin Med Phys
Phys Rev 98:1517–1518. 14:212–221.
Kao, Y. et al. (2013). Post-radioembolization Nickles, R.J. et al. (2004). Assaying and PET
yttrium-90 PET/CT—Part 2: Dose-response imaging of yttrium-90: 1>>34 ppm>0. IEEE
and tumor predictive dosimetry for resin Nucl Sci Symp Rec 6:3412–3414.
microspheres. EJNMMI Res 3:57. Okuda, K. et al. (1985). Natural history of hepa-
Kao, Y. et al. (2012). Yttrium-90 internal pair tocellular carcinoma and prognosis in relation
production imaging using first generation to treatment study of 850 patients. Cancer
PET/CT provides high-resolution images for 56:918–928.
qualitative diagnostic purposes. Br J Radiol Park, M., Mahmood, A., Zimmerman, R.,
85:1018–1019. Limpa-Amara, N., Makrigiorgos, G., Moore,
Kossert, K. et al. (2016). Comparison of 90Y S. (2008). Adsorption of metallic radionu-
activity measurements in nuclear medicine in clides on plastic phantom walls. Med Phys
Germany. Appl Radiat Isot 109:247–249. 35:1606.
Langhoff, H., Hennies, H. (1961). Zum experimen- Pasciak, A., Bourgeois, A., Bradley, Y. (2014a).
tellen Nachweis von Zweiquantenzerfall beim A comparison of techniques for 90Y PET/CT
0+–0+ Ubergang des Zr90. Z Phys 164:166–173. image-based dosimetry following radioem-
Lewellen, T. (1998). Time-of-flight PET. Semin bolization with resin microspheres. Front
Nucl Med 28:268–275. Oncol 4:121.
Lhommel, R., Goffette, P., Van den Eynde, M., Pasciak, A. et al. (2014b). Radioembolization and
Jamar, F., Pauwels, S., Bilbao, J., Walrand, the dynamic role of 90Y PET/CT. Front Oncol
S. (2009). Yttrium-90 TOF PET scan dem- 4:38.
onstrates high-resolution biodistribution Peller, P., Subramaniam, R., Guermazi, A. (2012).
after liver SIRT. Eur J Nucl Med Mol Imaging PET-CT and PET-MRI in Oncology. Berlin:
36:1696–1696. Springer.
Lhommel, R. et al. (2010). Feasibility of 90YTOF Rahmim, A. et al. (2005). Statistical dynamic
PET-based dosimetry in liver metastasis image reconstruction in state-of-the-art
therapy using SIR-spheres. Eur J Nucl Med high resolution PET. Phys Med Biol
Mol Imaging 37:1654–1662. 50:4887–4912.
References 249

Selwyn, R. et al. (2007). A new internal pair pro- Walrand, S. et al. (2015). The impact of image
duction branching ratio of 90Y: The develop- reconstruction bias on PET/CT 90Y dosim-
ment of a non-destructive assay for 90Y and etry after radioembolization. J Nucl Med
90Sr. Appl Radiat Isot 65:318–327. 56:494–495.
Stabin, M.G., Eckerman, K.F., Ryman, J.C., Wissmeyer, M. et al. (2011). 90Y time-of-flight
Williams, L.E. (1994). Bremsstrahlung radia- PET/MR on a hybrid scanner following liver
tion dose in yttrium-90 therapy applications. radioembolisation (SIRT). Eur J Nucl Med Mol
J Nucl Med 35:1377–1380. Imaging 38:1744–1745.
Sunderland, J., Christian, P., Kiss, T. (2015). PET/ Werner, M. et al. (2009). PET/CT for the assess-
CT scanner validation for clinical trials-reasons ment and quantification of 90Y biodistribution
for failure, recipes for success: The Clinical after selective internal radiotherapy (SIRT) of
Trials Network (CTN) experience. J Nucl Med liver metastases. Eur J Nucl Med Mol Imaging
56(3):1737. 37:407–408.
Surti, S. (2014). Update on time-of-flight PET Werner, M. et al. (2010). PET/CT for the detection
imaging. J Nucl Med 56:98–105. and quantification of the β-emitting thera-
Surti, S., Karp, J. (2016). Advances in time-of- peutic radionuclide yttrium-90 after liver SIRT
flight PET. Phys Med 32:12–22. doi: 10.1016/j. [abstract]. J Nucl Med 51(Suppl 2):341.
ejmp.2015.12.007. Willowson, K. et al. (2012). Quantitative 90Y
Tapp, K. et al. (2014). The impact of image recon- image reconstruction in PET. Med Phys
struction bias on PET/CT 90Y dosimetry after 39:7153.
radioembolization. J Nucl Med 55:1452–1458. Willowson, K., Tapner, M., Bailey, D. (2015). A
Thiam, C., Bobin, C., Lourenço, V., Chisté, V., multicentre comparison of quantitative 90Y
Amiot, M., Mougeot, X., Lacour, D., Rigoulay, PET/CT for dosimetric purposes after radio-
F., Ferreux, L. (2016). Investigation of the embolization with resin microspheres. Eur J
response variability of ionization chambers for Nucl Med Mol Imaging, 42:1202–1222.
the standard transfer of SIR-Spheres®. Appl Woods, M. et al. (1996). Calibration of the NPL
Radiat Isotopes 109:231–235. secondary standard radionuclide calibrator
van Elmbt, L. et al. (2011). Comparison of for 32P, 89Sr and 90Y. Nucl Instrum Methods
yttrium-90 quantitative imaging by TOF and Phys Res A 369:698–702.
non-TOF PET in a phantom of liver selec- Zimmerman, B., Ratel, G. (2005). Report of
tive internal radiotherapy. Phys Med Biol the CIPM key comparison CCRI(II)-K2Y-90.
56:6759–6777. Metrologia 42:06001.
12
Image-based three-dimensional
dosimetry following radioembolization

ALEXANDER S. PASCIAK AND S. CHEENU KAPPADATH

12.1 Introduction 251 12.5 LDM for image-based epatic dosimetry 255
12.2 Fully 3D Monte Carlo transport for 12.5.1 Validation of the LDM 256
image-based hepatic dosimetry 252 12.5.2 LDM and alternative
12.3 Dose-point kernel convolution for radionuclides 258
image-based hepatic dosimetry 253 12.6 Instances where DPK convolution and
12.4 The voxel S-value MIRD approach for LDM may not be appropriate 259
image-based hepatic dosimetry 254 12.7 Conclusion 260
References 260

follow-up. For example, if radiation dose and,


12.1 INtrODUCtION therefore, toxicity to normal or extrahepatic
tissues can be determined immediately follow-
The general dosimetric principles of 90Y radio- ing radioembolization, prompt administration
embolization have been discussed in detail of prophylaxis can be considered which may
in several chapters in this book. For example, decrease the severity of side effects. Furthermore,
Chapter  5 discusses a commonly used formula undertreated areas of tumor could be identified
that allows for the calculation of the absorbed and alternative or adjuvant therapies could be
dose to a volume of tissue given a uniform dis- prescribed, increasing the potential efficacy of
90Y radioembolization in some patients.
tribution of 90Y activity. While this is useful for
determining the average absorbed dose to the The ability to utilize posttreatment dosimetry
liver, lobe, or segment treated, it is of limited in the aforementioned manner depends on the
use in the estimation of biological effect. More availability of several important pieces of data.
accurate methods for determining absorbed dose First, one must understand the radiation biology
have been suggested such as the partition model, and dose–response properties of the normal liver
discussed in Chapters 4 and 5. However, the par- and the tumor, which may vary substantially with
tition model cannot account for inhomogeneity tumor size, type, and other factors. The radiation
of microsphere distribution in tumor or normal biology of radioembolization at a macroscopic and
liver. Determining the absorbed dose following a microscopic level has been discussed in Chapters
90Y radioembolization in all areas of the tumor, 8 and 9, respectively. However, there are still many
uninvolved liver, and extrahepatic tissues is a unknowns yet to be addressed. Once the radiation
critical future component of managing patient biology is understood for a patient with a given

251
252 Image-based three-dimensional dosimetry following radioembolization

history, the second missing piece of data is a com- In this chapter, we will discuss the four primary
plete description of the absorbed dose following methodologies for 90Y image-based dosimetry fol-
treatment. Postradioembolization three-dimen- lowing radioembolization.
sional (3D) image-based dosimetry is already
capable of providing this information in a routine
clinical scenario. 12.2 FULLY 3D MONtE CarLO
Chapters 10 and 11 have laid out the principles traNSPOrt FOr
behind 90Y posttreatment imaging using brems-
IMaGE-BaSED HEPatIC
strahlung single-photon emission computed
tomography (SPECT)/computed tomography
DOSIMEtrY
(CT) or 90Y positron emission tomography (PET)/
CT with an emphasis on quantification. However, A brief introduction to the mathematics of the
dosimetric calculation for internal emitters based Monte Carlo method has been included in Chapter
on SPECT or PET imaging is not a new idea—it 9. Monte Carlo methods are the de facto standard
was suggested long before “quantitative imaging” for radiation transport in medicine and are used
was a routine component of clinical vocabulary extensively in radiation oncology and radiology,
(Loevinger et al., 1989; Bolch et al., 1999). It is both clinically and in research. Even with modern
now standard practice to perform compartment- advancements in computer technology, the com-
based dosimetry for radiolabeled agents in vivo putational burden associated with fully 3D Monte
using SPECT or PET. Given a single injected Carlo simulation is significant. Its advantage, how-
bolus of a radioactive agent, pharmacokinetic ever, is that accurate dosimetric calculations can be
parameters can describe a complex relationship performed for radiation traversing any heteroge-
of uptake followed by clearance into the tissue neous material structure. If this material structure
of interest. Increasing this complexity, the time– is a patient, voxelized patient-specific phantoms
activity curve can vary from patient to patient, from CT image sets can be incorporated into the
especially in cancer therapy necessitating a Monte Carlo simulation. However, the utility of
patient-specific evaluation. As a result, dosimetry this for radioembolization is limited.
typically involves imaging at multiple time points When radiation penetrates a patient, whether it
in order to determine the activity concentration, is x-ray, γ-ray, or β-radiation, it does not discern
as a function of time, in both the tissue of interest among tissue types as one might initially think.
and the surrounding tissues. Radiation will interact differently in different tis-
Fortunately, serial imaging is not required sue types if there is (1) a substantial difference in
for image-based dosimetry following 90Y radio- electron density (electrons/cm3) or (2) a substantial
embolization. All currently used radioemboliza- difference in the atomic number of the atoms in
tion products are unique from pharmacologic the tissue. Unlike almost everything else in medi-
radioactive agents in that they are brachytherapy cine, the behavior of radiation is unaffected by bio-
devices, not drugs. As previously mentioned, chemical structure. Radiation will travel through
both glass and resin 90Y microspheres, as well as healthy tissue, diseased tissue, and venous/arterial
166 Ho microspheres, form a nonbiodegradable tissue in exactly the same way. Even adipose tis-
permanent implant upon infusion, where they sue has little effect on radiation from a dosimet-
release their radiation burden locally (Chapter 1). ric standpoint, less a small change in density. In
As discussed in Chapter 7, little or no systemic fact, accurate transmission of radiation through a
release of 90Y from the microspheres is seen in patient can be performed by lumping the patient’s
vivo. A single-session posttreatment quantitative tissues into three broad categories: soft tissue,
scan via either bremsstrahlung SPECT/CT or 90Y bone tissue, and lung tissue. Since the liver is one
PET/CT can be used to compute the committed of the most physically homogeneous organs in the
absorbed dose of the 90Y as it decays. Of course, body, the advantage to using Monte Carlo radia-
the limitations of the imaging system (resolu- tion transport methods is, therefore, lost in most
tion, quantification accuracy, and noise) will be cases. For this reason, other methods for image-
directly translated into errors in the 3D dose based dosimetry following 90Y radioembolization
map. are often employed.
12.3 Dose-point kernel convolution for image-based hepatic dosimetry 253

less of an issue for two reasons. First, because of


12.3 DOSE-POINt KErNEL the limited resolution of SPECT and PET, A(x,y,z)
CONVOLUtION FOr matrix sizes are usually small. Further, unlike
IMaGE-BaSED HEPatIC gamma emitters, 90Y β emission has a limited
DOSIMEtrY range and the DPK does not need to cover more
than 11 mm for any single octant. Therefore, the
Multiple authors have discussed application small matrix sizes defining A and DPK permit
of dose-point kernel (DPK) convolution as an relatively fast 3D convolution.
approach to absorbed-dose determination in vox- By convention, a 90Y DPK is computed using a
elized phantoms (Bolch et al., 1999; Strigari et al., validated Monte Carlo code for a predefined voxel
2006; Pasciak and Erwin, 2009; Kennedy et al., size. 90Y source specification is often uniformly
2011; D’Arienzo et al., 2013; Elschot et al., 2013). distributed through the origin voxel, sometimes
DPK convolution was desirable in past decades with the inclusion of microsphere composition and
since Monte Carlo-based transport, particularly size (Paxton et al., 2012). 90Y β spectra have been
for high-energy electrons, carried a substantial reported by a number of sources and are avail-
computation burden. Of course, because of the able from Eckerman et al. (1994) with energy bins
inherent 3D homogeneity of liver tissue combined beginning at 0.1 keV up to the maximum energy
with the limited range of 90Y β emissions, there of the emission. DPKs are computed for a single
is a little difference between DPK convolution transformation of the radionuclide of interest,
and voxel-based fully 3D Monte Carlo transport necessitating additional scaling factors to convert
(Pasciak and Erwin, 2009). Dome lesions at the the activity concentration matrix, A, into the total
apex of the liver may be a potential exception to number of transformations in each voxel, inte-
this general rule (Mikell et al., 2015). Image-based grated from time 0 to infinity. This is accomplished
dosimetry following 90Y posttreatment imaging with the 1/λ constant in Equation 12.1.
using a 90Y DPK is defined as follows: In general, any Monte Carlo code capable of
3D transport of electrons through voxel geome-
tries can be used to compute a DPK. The majority
1 of modern Monte Carlo codes use a condensed
D( x , y , z ) = ( A ⊗ DPK)( x , y , z )
λ history approximation to decrease the sizeable
computational burden associated with track-
∑ ∑ ∑ A(x ′, y ′, z ′)⋅
1 (12.1)
= structure electron transport simulation. Using
λ z′
x′ y′
the condensed history method, the combined
DPK( x − x ′ , y − y ′ , z − z ′ ) energy losses along the track from multiple inter-
actions (including excitation and electron impact
where λ is the decay constant of the radionu- ionization) are summed into a larger pseudoin-
clide and A(x,y,z) is the 3D activity concentration teraction (Berger, 1963). While there are some
matrix determined from quantitative imaging. instances where the condensed history tech-
While more computationally efficient than 3D nique cannot provide sufficient accuracy, these
Monte Carlo simulation, convolution of the activ- instances are not found in the computation of 90Y
ity concentration matrix, A, by the DPK can still DPKs for clinical hepatic dosimetry. The follow-
be computationally demanding in certain cir- ing codes have been used for 90Y DPK determina-
cumstances. Mathematical transformations can tion and point dosimetry: GEANT4 (Pacilio et al.,
ease this burden, such as the fast Hartley trans- 2009; Guimarães et al., 2010), MCNP5 (Paxton
form, which is an integral transformation simi- et  al.,  2012), MCNP4C (Pacilio et al., 2009),
lar to Fourier methods, except that it eliminates MCNPX (Dieudonne et al., 2010), EGSNRC
complex solutions. Following Fourier or Hartley (Strigari et al., 2006; Pacilio et al., 2009; Paxton
transformation, multiplication of A and DPK in et al., 2012), and EGS4 (Strigari et al., 2006);
the frequency domain carries equivalence to con- however, this is not an exhaustive list. Although
volution with a substantially decreased compu- not specific to 90Y, some detailed analyses of the
tational burden. However, for DPK convolution differences in electron transport dose profiles
applied to 90Y image-based dosimetry, speed is with codes utilizing different electron transport
254 Image-based three-dimensional dosimetry following radioembolization

algorithms have been published (Uusijarvi et al.,


2009). While the aforementioned manuscript has 12.4 tHE VOXEL S-VaLUE
found minor variations in the results of different MIrD aPPrOaCH FOr
Monte Carlo codes for 90Y dosimetry, the clini- IMaGE-BaSED HEPatIC
cal importance of these differences should be put DOSIMEtrY
into perspective. After reading the discussion on
the local deposition method (LDM) later in this The voxel S-value (VSV) Medical Internal
chapter, it will become apparent that minor dif- Radiation Dose (MIRD) approach to image-based
ferences in DPK are likely to carry a negligible dosimetry is an alternative to DPK convolution
clinical impact. Instead, a Monte Carlo code with a formalism analogous to MIRD S-values,
which the user is comfortable with and finds traditionally used for organ → organ dosimetry
easy to use should be selected. For example, the of internal emitters. The VSV method was first
authors of this chapter find EGSNRC to be sub- described by Bolch et al. (1999). The relationship
stantially easier to use than EGS4. for determining the absorbed dose in target voxel
The majority of published 90Y DPKs are for (DVoxelT) using VSVs is given in
liver tissue only. Although not generally a part
of routine clinical 90Y radioembolization, there
may be some scenarios where 3D image-based
DVoxel T =
1
λ ∑A Voxel S ⋅ S ( Voxel T ← VoxelS ) (12.2)
N
lung dosimetry might be performed using DPK
convolution. For example, such calculations may where S is the average energy deposition in the
be useful for root cause analysis following the target voxel, VoxelT, from a single transformation
occurrence of clinical side effects such as radia- in the source voxel, VoxelS. Corresponding to the
tion pneumonitis after treatment. In these cases, maximum range of a 90Y β-emission, VSVs for
the Monte Carlo calculation of lung DPKs can be all source voxels (N) within 11 mm of the target
computed. Alternatively, one can perform a first- voxel should be summed. As with DPK convolu-
order correction to existing DPKs for liver tissue tion, the 1/λ constant converts activity in a voxel to
using density scaling to account for differences the total number of transformations. While DPKs
between lung and soft tissue. Mikell et al. (2015) are stored and applied in 3D matrix form, S-values
discuss this further. are tabulated for each target voxel as a function of
In the previous chapters of this book, it has source voxel location.
been emphasized that 90Y is predominantly a pure The VSV MIRD approach is defined differently
β emitter. However, high-energy β particles gen- than DPK convolution; however, many properties
erate bremsstrahlung x-rays as they slow down in are identical. Like DPK convolution, inhomoge-
an absorber, creating a coupled photon–electron nous tissues cannot be accommodated that, again,
spectrum within the patient. Bremsstrahlung is generally not an issue for hepatic 90Y radioem-
x-rays have the propensity to penetrate through bolization. Further, VSVs and DPKs are intercon-
substantially greater tissue thickness than 90Y β vertible and when applied to the same activity
particles, which may seem to confound 3D dosim- concentration matrix will yield the same dosimet-
etry. Certainly, coupled photon–electron transport ric solution. An advantage in the computational
using Monte Carlo methods can be performed with simplicity of the VSV method compared with DPK
high accuracy. However, the dose contribution of convolution can be realized when calculating the
bremsstrahlung photons will not be accounted for absorbed dose to a subset of voxels in the volume,
when using 90Y DPKs, which cover only the maxi- for example, to calculate isodose curves. However,
mum β range. However, as demonstrated by Stabin when computing the absorbed dose at every voxel
et al. (1994), the hepatic absorbed dose contribu- (i.e., in the entire 3D image set), the computational
tion from bremsstrahlung x-rays is three orders burden of the VSV MIRD approach is equivalent
of magnitude below that of the 90Y β emission. to that of DPK convolution.
Therefore, bremsstrahlung x-rays will have a neg- One problem with both VSVs and DPK convolu-
ligible impact on the accuracy of DPK and other tion is that in a modern clinical environment, voxel
non-Monte Carlo methods for 3D dosimetry dis- size often varies with both scanner and image recon-
cussed in the remainder of this chapter. struction parameters. Under normal circumstances,
12.5 LDM for image-based epatic dosimetry 255

this makes the routine clinical use of these tech- mathematics and computation associated with 3D
niques challenging since tabulated DPKs and VSVs postradioembolization dosimetry to a simple scal-
are available for only a limited array of 90Y voxel sizes. ing factor. The following derivation describes the
However, additional methods have been proposed for scaling factor used in the LDM.
quickly generating VSVs for an arbitrary sized voxel Under the aforementioned assumptions, the
from predetermined data. Fernandez et al. (2013) absorbed dose, D 90 Y (Gy), within a voxel can be
used Monte Carlo methods to perform electron determined according to
transport for several radionuclides, including 90Y at A0 (Bq / mL) ⋅ 4.998 × 10−8 (J ⋅ s)
a voxel pitch of 0.5 mm. These data have been made D 90 Y (Gy) = (12.3)
ρliver (kg / mL)
publically available; however, what is unique is their
proposed method to analytically rescale these data where A0 is the activity concentration of 90Y (Bq/
for use in problems with arbitrary voxel size, without mL) within the voxel and ρ is the density of liver
rerunning the Monte Carlo simulation. Fernandez et tissue in kg/mL. The constant factor 4.998 × 10 −8 J.s
al. has validated their rescaling method by compar- is the energy released per unit activity of 90Y, previ-
ing it to native Monte Carlo simulations performed ously derived in both Chapters 5 and 7. If the den-
for the following voxel sizes: 0.5, 0.7, 1.0, 1.23, 1.5, 1.8, sity of liver tissue as defined by the International
2.0, 2.4, 3.0, 4.8, 6.0, 8.0, and 10.0 mm. When used Commission on Radiation Units and Measures,
for image-based dosimetry, the Fernandez rescaling ICRU (1992) is selected, the constants in Equation
method introduces less than 1.5% error compared 12.3 can be grouped into a single conversion factor
with calculating VSVs directly with the Monte Carlo K90Y, valid only for 90Y. Note that Equation 12.3 is
method (Fernández et al., 2013). To this end, several independent of voxel volume. This powerful fea-
other authors have suggested methods for on-the-fly ture allows LDM to be utilized regardless of recon-
rescaling of VSVs for use in image-based dosimetry struction matrix size or isotropicity following
that are worthy of consideration (Dieudonne et al., bremsstrahlung SPECT/CT or 90Y PET/CT post-
2011; Amato et al., 2012). treatment imaging. However, the accuracy of LDM
As previously mentioned, the dosimetric solu- still rests on the assumption that the entirety of the
tions computed from both the VSV MIRD approach energy from each decay is absorbed by the voxel
and DPK convolution will be equivalent. The ensu- where the decay occurred. This assumption may be
ing discussions will compare the accuracy of alter- violated as voxel volume decreases or as charged-
native computation techniques to DPK convolution, particle equilibrium is lost between voxels. This
and it should be understood that these comparisons will be discussed in detail in Section 12.5.1.
apply to the VSV MIRD method as well.
 Bq   mL 
D 90 Y (Gy) = A0   ⋅ K 90 Y  Gy ⋅ (12.4)
 mL   Bq 
12.5 LDM FOr IMaGE-BaSED
Equation 12.4 is identical to Equation 12.3,
EPatIC DOSIMEtrY except that constants have been replaced by the
K90Y factor previously mentioned. For any quan-
The LDM is the final method that we will dis- titative 90Y image set, with units of 90Y activity
cuss for performing 90Y postradioembolization concentration at the time of microsphere infusion,
image-based dosimetry. As with prior techniques, a K90Y factor of 4.782 × 10 −5 (Gy-mL/Bq) can be
quantitative postradioembolization imaging from used to determine the absorbed dose in every voxel
either bremsstrahlung SPECT/CT or 90Y PET/CT using the LDM.
is required as a starting point for dosimetry. Image As described in Chapter 10, some work is
sets must be quantified to activity concentration required to quantify 90Y SPECT/CT images
(Bq/mL) of 90Y at the time of infusion in order to using bremsstrahlung SPECT/CT; therefore, it
perform dosimetry using the LDM. is expected that as part of this effort appropri-
The LDM is based on the premise that the ate activity concentration units (Bq/mL of 90Y)
entirety of the energy released from each decay will be determined. However, in many cases 90Y
of 90Y within a voxel is deposited locally, within PET/CT will produce quantitative posttreatment
that same voxel. This assumption reduces all images without any additional effort by the end
256 Image-based three-dimensional dosimetry following radioembolization

user. As described in Chapter 11, this is often true  mL   mL  BX


even if the PET/CT manufacturer never intended K x  Gy ⋅  = K 90 Y  Gy ⋅ ⋅ (12.6)
 Bq   Bq  B90 Y
direct imaging of 90Y. As a result, when scanning
a patient postradioembolization using PET/CT
90Y may not be an available option on the scanner λ c = λ 90 Y − λ X (12.7)
console, although several PET/CT manufacturers
have made an effort to include it on recent soft- Equations 12.6 and 12.7 illustrate the deriva-
ware updates. tion of K x and λc for radionuclide X based on the
At a basic level, quantification for differ- relative branching ratio, β, and decay constant, λ.
ent positron-emitting radionuclides by PET/CT Values for K X and λc have been computed for 22Na
systems is performed with a simple rescaling of and 68Ge in Table 12.1. Note that K X is equivalent to
pixel values based on the half-life and branch- K 90 Y K90Y in Table 12.1.
ing ratio for positron emission of the injected
nuclide. Therefore, this process can easily be
integrated into the K scaling factor in Equation 12.5.1 VALIDATION OF THE LDM
12.4 for systems that do not directly support 90Y.
This is shown in Equation 12.5: As previously mentioned, the LDM rests upon the
assumption that all energy released in the decay
of radioactivity within a voxel contributes to the
 Bq   mL  λc ⋅t
D 90 Y (Gy) = A0  ⋅ K Gy ⋅ ⋅ e (12.5) absorbed dose in the same voxel. Several authors
 mL  x  Bq  have utilized LDM for image-based 90Y dosim-
etry. An indirect approach was explored by Chiesa
If 90Y is not directly supported by the PET/CT et al. (2012) and Mazzaferro et al. (2013), who both
system in question, but phantom experiments with used Tc-99m macroaggregated albumin (99mTc-
90Y suggest accurate quantification (Chapter 10),
MAA) to perform dosimetry based on LDM. Use
the LDM can still be used. Scanning a postradio- of LDM with 99mTc-MAA SPECT/CT was first sug-
embolization patient in these instances as either gested by Pasciak and Erwin (2009). While LDM
22Na or 68Ge will also allow for posttreatment
is fully capable of producing 3D dosimetry based
dosimetry of 90Y under the above conditions with on 99mTc-MAA, its accuracy rests on the additional
the LDM. K x in Equation 12.5 is a modified scaling assumption that the relative activity distribution of
factor that includes the branching ratio for posi- MAA mirrors that of 90Y radioembolization. This
tron emission of the radionuclide selected on the assumption may or may not be true as discussed in
scanner (i.e., 22Na or 68Ge) relative to the branching Chapters 10 and 11.
ratio of 90Y. The exponential term and the adjusted LDM has been used clinically for dosimetry by
decay constant (λc) accounts for the difference in several authors following 90Y PET/CT (Kao et al.,
half-life between 90Y and the radionuclide selected. 2013; Bourgeois et al., 2014; Srinivas et al., 2014)
However, for posttreatment imaging obtained on and its use has been validated specifically for post-
the day of treatment, the exponential can be omit- radioembolization imaging based on PET (Pasciak
ted with minimal introduced error so long as 22Na et al., 2014) and SPECT (Pacilio et al., 2015). One
or 68Ge are used: important consideration that is often ignored in

Table 12.1 90 Y dosimetry using PET/CT and the local deposition method
Branching ratio adjusted decay Conversion
Isotope used for for positron Decay constant λ constant λc factor KX (Gy-mL/
imaging emission (hours−1) (hours−1) Bq)
90Y 0.000032 1.083 × 10−2 0 4.782 × 10−5 (K90Y)
22Na 0.905 3.038 × 10−5 1.080 × 10−2 1.353
68Ge 0.890a 1.066 × 10−4 1.072 × 10−2 1.330
a Adapted from Pasciak, A.S. et al. (2014). Front Oncol 4:121. doi: 10.3389/fonc.2014.00121. Branching ratio of
daughter, 68Ga, which is in secular equilibrium with the parent radionuclide, 68Ge.
12.5 LDM for image-based epatic dosimetry / 12.5.1 Validation of the LDM 257

postradioembolization image-based dosimetry  is estimate Dtrue than DPK convolution. Under ideal
that the bremsstrahlung SPECT or 90Y PET/CT circumstances and in the absence of image noise,
images used as a starting point are not perfect the accuracy of LDM and DPK can be directly
representations of the true activity concentration. If a compared as a function of scanner resolution,
90Y imaging technique is said to be quantitative, then defined by PSF at FWHM. Pasciak et al. performed
it can be used to determine the activity concentra- this ideal mathematical comparison, as well as one
tion at the center of a large phantom homogeneously based on phantom data with a particular focus
filled with 90Y. However, the activity concentration on 90Y PET/CT (Pasciak et al., 2014). The ideal
of 90Y microspheres in tumor is not homogeneous approach involves a precise mathematical phan-
and because of the imperfect resolution of the imag- tom convolved with a range of 3D Gaussian blur
ing method, the true activity concentration will be kernels to simulate the shift-invariant PSF repre-
convolved (blurred) by the point-spread function sentative of either 90Y PET/CT or SPECT. For the
(PSF) of the imaging modality: results in Figure 12.2, the mathematical phantom
simulated was the NEMA IEC body phantom,
A ( x , y , z ) = T ( x , y , z ) ⊗ PSF (12.8) with hot spheres ranging from 10 to 37 mm in size.
Gaussian kernels with FWHM ranging from 2.0 to
where A is the 3D activity concentration reported 15.0 mm in increments of 0.25 mm were applied to
by quantitative imaging and T is the true 3D activ- create simulated activity concentrations, A(x,y,z).
ity concentration. If we revisit Equation 12.1 where Either DPK convolution or the LDM was applied
we defined the relationship used for DPK convo- to A(x,y,z) for subsequent comparison with the
lution in image-based 3D dosimetry, we can now Dtrue(x,y,z). In this case, since T(x,y,z) is known,
see that it contains an inaccuracy. The inaccuracy Dtrue can be computed exactly as shown in Equation
in Equation 12.1 is that A(x,y,z) is convolved by 12.9. Figure 12.2 shows the results of a voxel-by-
the DPK. In actuality, the true 3D absorbed dose voxel comparison of absorbed dose using the LDM
(Dtrue) is described in: and DPK convolution in three of the phantom hot
spheres as a function of the scanner PSF. It can be
1
λ
(T ( x , y , z ) ⊗ DPK ) (12.9)
Dtrue ( x , y , z ) =  seen in Figure 12.2 that LDM has the best accuracy
with a scanner FWHM of just over 4 mm, where
Unfortunately, because T(x,y,z) cannot be the PSF closely approximates the shape of the 90Y
determined in a patient, the aim is to find the best DPK. At a FWHM of less than 4 mm, the accuracy
method for approximating Dtrue given the reported of LDM worsens while the accuracy of DPK convo-
activity concentration, A(x,y,z). lution continues to improve until perfect scanner
The PSF measured at full-width and half- resolution is obtained (FWHM = 0 mm). However,
maximum (FWHM) for 90Y PET/CT ranges for PSF FWHM greater than 4 mm, LDM always
between 3.1 and 10.5 mm, depending on the scan- carries a slight advantage. It should be noted that
ner and reconstruction parameters, as discussed in the shape of the PSF and reconstructed voxel size
Chapter 11. For bremsstrahlung SPECT, expected will affect the relationships in Figure 12.2.
PSFs will be substantially higher than 90Y PET/ Any accuracy gained with the use of LDM
CT as discussed in Chapter 10. However, because depends on image reconstruction parameters, scan-
of the relatively poor resolution of both 90Y brems- ner PSF, voxel size, image noise, and also, patient
strahlung SPECT and 90Y PET, the blur in the data motion. Several authors have investigated some or
introduced by the scanner PSF is often greater all of these factors and their effect on the accuracy
than the spread in energy deposition computed of LDM relative to DPK convolution for both 99mTc-
by convolution with the DPK. Therefore, in some MAA SPECT and 90Y bremsstrahlung SPECT-based
circumstances use of the LDM, which introduces dosimetry (Pasciak and Erwin, 2009; Ljungberg and
no additional blur, may result in an absorbed dose Sjögreen-Gleisner, 2011; Pacilio et al., 2015) as well
map that is closer to Dtrue than the absorbed dose as 90Y PET/CT (Pasciak et al., 2014). A common
map calculated using DPK convolution. This is consensus between all of these reports is that with
qualitatively illustrated in Figure 12.1. the current 90Y imaging technology, either brems-
Indeed, the qualitative example in Figure strahlung SPECT or 90Y PET/CT, neither DPK con-
12.1 suggests that LDM might more accurately volution nor the VSV MIRD formalism provides
258 Image-based three-dimensional dosimetry following radioembolization

(a) (b)

T(x,y,z)
T(x,y,z) Ⓧ DPK Dtrue(x,y,z)

T(x,y,z) Ⓧ PSF

(c) (d)

A(x,y,z) Ⓧ DPK

A(x,y,z)

Figure 12.1 A qualitative example illustrating the effect of scanner PSF and DPK convolution on the
shape of the absorbed dose profile for a point source of 90Y occupying a single voxel. The voxel size in
this example is 2.0 mm3. The scanner PSF is approximated by a 6.5-mm FWHM Gaussian filter, which
has been shown to be a reasonable approximation for 90Y PET/CT (Pasciak et al., 2014). (a) 2D repre-
sentation of a point source of 90Y activity, T(x,y,z). This represents the true activity distribution, which
is unknown for in vivo patient imaging; (b) convolution of the true activity concentration with the 90Y
DPK gives the true absorbed-dose distribution, Dtrue. This is also unknown for in vivo patient imag-
ing. (c) The true activity concentration convolved by the PSF of the imaging system produces A(x,y,z).
Since LDM applies no additional burring to A(x,y,z), the shape in (c) is the shape of the absorbed-dose
profile if the LDM is applied. (d) A(x,y,z) is convolved by the 90Y DPK. Notice the additional spread
in the absorbed-dose profile in (d) compared with (c). Both overestimate the spread of the true
absorbed-dose profile (Dtrue), owing to the 6.0-mm FWHM PSF of the imaging system in this fabri-
cated example.

an accuracy advantage over LDM. While it is pos- its ability to be directly imaged by SPECT/CT and
sible the LDM may produce slightly more accurate MRI. These characteristics have been reviewed in
results, this increased accuracy may not be clinically Chapters 1 and 15. 166Ho decays with the emission
detectible. In light of the fact that the LDM can be of an 80.5 KeV γ-ray with a branching ratio of 6%
applied with a simple scaling factor, it is highly rec- in addition to its therapeutic β emission. While
ommended for routine 90Y image-based dosimetry. one might initially assume this γ emission would
preclude the use of LDM since it will result in dose
deposition beyond the voxel of interest, the γ emis-
12.5.2 LDM AND ALTERNATIVE sion is unlikely to be of significance. Both Traino
RADIONUCLIDES et al. (2013) and Pacilio et al. (2015) previously
showed the validity of the use of the LDM for both
While this book has been primarily focused on 90Y 90Y and 131I. Owing to the much larger branching

radioembolization, 166Ho radioembolization may ratios of γ-emissions in 131I (81.5% for the 364.5
become more widespread in the future owing to keV γ-ray) compared with 166Ho, it is likely that the
12.6 Instances where DPK convolution and LDM may not be appropriate 259

40

% error with Dtrue (x,y,z)


30

20

10

0
2 4 6 8 10 12 14 16
Ideal imaging system PSF (mm) at full-width half-max
(a)
40
% error with Dtrue (x,y,z)

30

20

10

0
2 4 6 8 10 12 14 16
Ideal imaging system PSF (mm) at full-width half-max
(b)
40
Local deposition method (LDM)
% error with Dtrue (x,y,z)

DPK convolution
30

20

10

0
2 4 6 8 10 12 14 16
Ideal imaging system PSF (mm) at full-width half-max
(c)

Figure 12.2 Error analysis of DPK convolution and LDM in comparison to D true for mathematically
simulated NEMA IEC body phantom, in the absence of image noise. Error is presented as a function
of scanner PSF FWHM, with blur applied using a Gaussian kernel with a 2-mm isotropic voxel size.
Every voxel in each hot sphere was compared with corresponding voxels in D true, with average error,
maximum, and minimum indicated with the central, upper, and lower bounds of the shaded regions,
respectively. (a) 10 mm hot sphere; (b) 17 mm hot sphere; and (c) 37 mm hot sphere.

validity of LDM would also be extended to 166Ho; to the homogeneity of liver tissue. However, the
however, this has not been explicitly explored. inhomogeneous material density at the dome
of the liver represents one case where the uni-
formity requirement of DPK may not be valid.
Mikell et al. (2015) simulated the accuracy of
12.6 INStaNCES WHErE DPK
the aforementioned image-based dosimetry
CONVOLUtION aND LDM
techniques, including 3D Monte Carlo at the
MaY NOt BE aPPrOPrIatE liver–lung interface as a function of scanner res-
olution. Mikell demonstrated that DPK convolu-
As discussed in Section 12.1, fully 3D Monte tion, LDM, and 3D Monte Carlo are within 10%
Carlo radiation transport calculations are rarely of the true liver absorbed dose when deeper than
used for 90Y hepatic radioembolization owing 12 mm from the liver–lung interface; however,
260 Image-based three-dimensional dosimetry following radioembolization

this distance is expected to increase as the scan- Bourgeois, A.C. et al. (2014). Intra-procedural
ner resolution becomes poorer. Similarly, DPK, 90Y PET/CT for treatment optimization of
LDM, and 3D Monte Carlo achieved similar 90Y radioembolization. J Vasc Interv Radiol
accuracy in the lung when deeper than 39 mm 25:271–275.
from the interface. Monte Carlo-based dosimetry Chiesa, C. et al. (2012). A dosimetric treatment
is required for the precise determination of liver planning strategy in radioembolization
or lung absorbed dose at the interface. of hepatocarcinoma with 90Y glass
microspheres. Q J Nucl Med Mol Imaging
56:503–508.
D’Arienzo, M. et al. (2013). Absorbed dose to
12.7 CONCLUSION lesion and clinical outcome after liver radio-
embolization with (90)Y microspheres: A case
Quantitative postradioembolization imaging is report of PET-based dosimetry. Ann Nucl
feasible in routine clinical scenarios as shown Med 27:676.
in Chapters 10 and 11. This has largely been Dieudonne, A. et al. (2010). Fine-resolution voxel
made possible not only by 90Y PET/CT, but also S values for constructing absorbed dose dis-
by advancements in techniques to quantify 90Y tributions at variable voxel size. J Nucl Med
bremsstrahlung SPECT. The major barrier related 51:1600–1607.
to patient-specific image-based dosimetry for Dieudonne, A. et al. (2011). Clinical feasibility of
90Y has always been the lack of broadly avail- fast 3-dimensional dosimetry of the liver for
able quantitative imaging. With these data, and treatment planning of hepatocellular carci-
combined with the fact that radioembolization noma with 90Y-microspheres. J Nucl Med
is a permanent implant obviating the need for 52:1930–1937.
serial imaging, transformation into 3D absorbed Eckerman, K.F., Westfall, R.J., Ryman, J.C.,
dose becomes trivial. Methods used to generate Cristy, M. (1994). Availability of nuclear decay
absorbed-dose maps can range in complexity; data in electronic form, including beta spec-
however, for many clinical scenarios the applica- tra not previously published. Health Phys
tion of a simple scaling factor (LDM) is all that is 67:338–345.
necessary for excellent results. With these tech- Elschot, M. et al. (2013). Quantitative compari-
niques now becoming firmly established, the son of PET and bremsstrahlung SPECT for
much more challenging problem of developing a imaging the in vivo yttrium-90 microsphere
standard-of-care protocol for applying these data distribution after liver radioembolization. Villa
toward the management of patient care can begin E, ed. PLoS One 8:e55742.
to be addressed. Fernández, M. et al. (2013). A fast method for res-
caling voxel S values for arbitrary voxel sizes
REFERENCES in targeted radionuclide therapy from a single
Monte Carlo calculation. Med Phys 40:082502.
Amato, E. et al. (2012). An analytical method for Guimarães, C.C., Moralles, M., Sene, F.F.,
computing voxel S values for electrons and Martinelli, J.R. (2010). Dose-rate distribution
photons. Med Phys 39:6808–6817. of 32P-glass microspheres for intra-arterial
Berger, M.J. (1963). Monte Carlo calculation of brachytherapy. Med Phys 37:532–539.
the penetration and diffusion of fast charged ICRU. (1992). Photon, Electron, Proton and
particles. In Methods in Computational Neutron Interaction Data for Body Tissues.
Physics, Vol. I, Adler, B., Fernbach, S., ICRU Report 46. Bethesda: International
Rotenberg, M. (Eds). New York, NY: Academic Commission on Radiation Units and
Press, 135–215. Measurements.
Bolch, W.E. et al. (1999). MIRD pamphlet No. 17: Kao, Y.H. et al. (2013). Post-radioembolization
The dosimetry of nonuniform activity distri- yttrium-90 PET/CT—Part 2: Dose–response
butions—Radionuclide S values at the voxel and tumor predictive dosimetry for resin
level. J Nucl Med 40:11S–36S. microspheres. EJNMMI Res 3:1–1.
References 261

Kennedy, A., Dezarn, W., Weiss, A. (2011). Patient Pasciak, A.S., Erwin, W.D. (2009). Effect of voxel
specific 3D image-based radiation dose esti- size and computation method on Tc-99m
mates for 90Y microsphere hepatic radioem- MAA SPECT/CT-based dose estimation for
bolization in metastatic tumors. J Nucl Med Y-90 microsphere therapy. IEEE Trans Med
Radiat Ther 2:1–8. Imaging 28:1754–1758.
Ljungberg, M., Sjögreen-Gleisner, K. (2011). Paxton, A.B., Davis, S.D., DeWerd, L.A. (2012).
The accuracy of absorbed dose estimates in Determining the effects of microsphere and
tumours determined by quantitative SPECT: A surrounding material composition on 90Y
Monte Carlo study. Acta Oncol 50:981–989. dose kernels using EGSnrc and mcnp5. Med
Loevinger, R., Kassis, A.I., Burt, R.W. (Eds.). (1989). Phys 39:1424–1434.
The MIRD Perspective. Washington, DC: The Srinivas, S.M. et al. (2014). Determination of
American College of Nuclear Physicians. radiation absorbed dose to primary liver
Mazzaferro, V., Sposito, C., Bhoori, S., Romito, R. tumors and normal liver tissue using post-
(2013). Yttrium‐90 radioembolization for inter- radioembolization (90)Y PET. Front Oncol
mediate‐advanced hepatocellular carcinoma: 4:255.
A phase 2 study. Hepatology 57:1826–1837. Stabin, M.G., Eckerman, K.F., Ryman, J.C.,
doi: 10.1002/hep.26014. Epub 2013 Mar 22. Williams, L.E. (1994). Bremsstrahlung radia-
Mikell, J.K. et al. (2015). Comparing voxel-based tion dose in yttrium-90 therapy applications.
absorbed dosimetry methods in tumors, J Nucl Med 35:1377–1380.
liver, lung, and at the liver-lung interface for Strigari, L., Menghi, E., D’Andrea, M., Benassi, M.
90Y microsphere selective internal radiation (2006). Monte Carlo dose voxel kernel calcula-
therapy. EJNMMI Phys 2:947. tions of beta-emitting and Auger-emitting
Pacilio, M. et al. (2015). Differences in 3D dose radionuclides for internal dosimetry: A com-
distributions due to calculation method parison between EGSnrcMP and EGS4. Med
of voxel S-values and the influence of Phys 33:3383.
image blurring in SPECT. Phys Med Biol Traino, A.C. et al. (2013). Dosimetry for nonuni-
60:1945–1964. form activity distributions: A method for the
Pacilio, M. et al. (2009). Differences among calculation of 3D absorbed-dose distribution
Monte Carlo codes in the calculations of voxel without the use of voxel S-values, point ker-
S values for radionuclide targeted therapy nels, or Monte Carlo simulations. Med Phys
and analysis of their impact on absorbed dose 40:042505.
evaluations. Med Phys 36:1543–1552. Uusijarvi, H. et al. (2009). Comparison of electron
Pasciak, A.S., Bourgeois, A.C., Bradley, Y.C. dose-point kernels in water generated by the
(2014). A comparison of techniques for Monte Carlo codes, PENELOPE, GEANT4,
90Y PET/CT image-based dosimetry fol- MCNPX, and ETRAN. Cancer Biother
lowing radioembolization with resin micro- Radiopharm 24:461–467.
spheres. Front Oncol 4:121. doi: 10.3389/
fonc.2014.00121.
13
Diagnostic reporting using
postradioembolization imaging

YUNG HSIANG KAO

13.1 Introduction 263 13.4.3 Nontarget activity versus noise 270


13.2 Bremsstrahlung SPECT/CT 264 13.4.4 Technical failure 272
13.3 90Y PET/CT 264 13.4.5 Quantification of nontarget
13.3.1 Advantages over absorbed dose 274
bremsstrahlung SPECT/CT 264 13.4.6 Verification of absorbed doses 275
13.3.2 Technical challenges of 90Y PET 265 13.5 Economics of 90Y PET 277
13.4 Principles of diagnostic reporting 268 13.6 Conclusions 278
13.4.1 Continuity of care 269 Acknowledgments 278
13.4.2 Target activity 269 References 278

The term “technical success” refers to whether


13.1 INtrODUCtION the treatment was carried out according to pre-
therapy planning expectations based on the quali-
As described in Chapter 1, yttrium-90 (90Y) radio- tative assessment of angiography and subsequent
embolization is a point-source brachytherapy microsphere biodistribution (Salem et al., 2011;
delivered by millions of radioactive microspheres Kao et al., 2013a). Technical success cannot be
permanently embedded within the target vascular determined by angiographic findings alone due to
territory and dispersed by intra-arterial injection. irreconcilable biophysical and injection technique
The aim of postradioembolization imaging is to differences between soluble angiographic contrast
verify the intended radiation plan. This consists of molecules versus particulate microspheres (Kao
two components: first to confirm technical success et al., 2011; Jiang et al., 2012). Technical success
and second to assess the likelihood of clinical suc- should not be confused with “clinical success,”
cess or serious toxicity based on tissue absorbed which takes into consideration the tissue effects of
doses. Postradioembolization imaging may be microsphere radiobiology as revealed on follow-up
achieved either indirectly using bremsstrah- imaging or biochemistry (Salem et al., 2011; Kao
lung single-photon emission computed tomog- et al., 2013a).
raphy with integrated computed tomography While the focus of this chapter is limited to
(CT) (SPECT/CT) or directly by positron emis- direct or indirect imaging of 90Y postinfusion,
sion tomography with integrated CT (PET/CT) of Chapter 14 will focus on imaging techniques used
minuscule positron emission. in follow-up posttherapy.

263
264 Diagnostic reporting using postradioembolization imaging

bremsstrahlung SPECT/CT, it may imply a clini-


13.2 BrEMSStraHLUNG cally significant amount of nontarget 90Y activ-
SPECt/Ct ity. Depending on the nontarget organ involved
and likelihood of complications, such cases may
Today’s minimum standard for postradioembo- require urgent clinical attention to mitigate poten-
lization imaging is bremsstrahlung SPECT/CT. tial toxicity. The general technique of nontarget
The advantage of SPECT/CT over planar scintig- activity assessment by bremsstrahlung SPECT/CT
raphy is the tomographic assessment of activity is similar to that developed for 90Y PET/CT (Table
biodistribution with the added specificity of CT 13.1 and Figure 13.2). The topic of nontarget toxic-
correlative anatomy and CT attenuation correc- ity risk assessment is further discussed in Section
tion. Planar bremsstrahlung scintigraphy of the 13.4.5 and in Chapter 14.
abdomen yields little clinically useful information Overall, bremsstrahlung SPECT/CT can only
and should be superseded by SPECT/CT. Even the provide a gross overview of the general micro-
visual assessment of hepatopulmonary shunting sphere biodistribution. It typically cannot con-
by planar bremsstrahlung scintigraphy is subjec- fidently confirm the presence or absence of
tive and of limited clinical value beyond what has activity within subcentimeter lesions or confi-
already been estimated by technetium-99m (99mTc) dently exclude nontarget activity, although there
macroaggregated albumin (MAA). are exceptions. Preliminary data using pinhole
As a form of scatter radiation of a continu- collimators for bremsstrahlung scintigraphy may
ous energy spectrum without a clear photopeak, partially ameliorate some of these problems and
bremsstrahlung scintigraphy is limited by inher- may benefit from further research (Walrand et
ently poor spatial resolution and quantitative inac- al., 2011). Currently, the qualitative and quantita-
curacy. These factors limit its clinical utility as a tive capabilities of bremsstrahlung SPECT/CT are
diagnostic modality for qualitative and quantita- suboptimal for diagnostic reporting and therefore
tive analyses of target and nontarget activity. Both an alternative modality for postradioembolization
bremsstrahlung planar and SPECT/CT images imaging should be sought.
appear blurry and are generally of low quality,
precluding reliable assessment of subcentimeter
lesions. This means that if the majority of targeted
tumors are small, technical success will be difficult 13.3 90Y PEt/Ct
to confirm. As a result, bremsstrahlung SPECT/
CT is frequently indeterminate for subcentimeter 13.3.1 ADVANTAGES OVER
lesions or nontarget activity (Kao et al., 2014a), BREMSSTRAHLUNG SPECT/CT
for example, a portal vein tumor thrombosis
(Figure 13.1). Furthermore, the thin visceral walls 90Y PET/CT is the most promising modality to
of the stomach, duodenum, and gallbladder are replace bremsstrahlung SPECT/CT due to its
often inseparable from the adjacent liver tissue on superior qualitative and quantitative capability.
bremsstrahlung SPECT/CT. Liver movement due Coincidence imaging of 90Y PET is possible due to
to tidal breathing may also lead to SPECT/CT mis- positron emission resulting from its natural decay
registration. Compounded together, all of these (Nickles et al., 2004). Conventional PET/CT or PET/
technical issues adversely affect the clinical utility magnetic resonance (MR) scanners with time-of-
of bremsstrahlung SPECT/CT for the assessment flight (TOF) may be used for coincidence imag-
of target and nontarget activity. ing of positrons from 90Y decay without hardware
Quantification of bremsstrahlung radiation modification (Lhommel et al., 2009; Wissmeyer
is technically challenging and largely inaccu- et al., 2011). 90Y PET/CT can obtain high-resolution
rate. Special techniques are required to quantify images of microsphere biodistribution with spatial
these images, as detailed in Chapter 10. However, resolutions between 5 and 10 mm (Pasciak et al.,
regardless of whether quantification methods 2014a). This means that 90Y PET has the capability to
have been employed, if a nontarget activity has assess for the presence or absence of activity within
unequivocally been detected qualitatively by subcentimeter lesions (Figure 13.3).
13.3 90 Y PET with integrated CT / 13.3.2 Technical challenges of 90Y PET 265

(a) (b)

(c) (d)

(e) (f )

Figure 13.1 Inferior vena cava tumor thrombosis in multifocal hepatocellular carcinoma (arrow). (a, b)
Triphasic computed tomography (CT) liver in the arterial phase in trans-axial and coronal planes; (c, d)
yttrium-90 positron emission tomography with integrated CT (90Y PET/CT) in trans-axial and coronal
planes depict focal activity within the inferior vena cava tumor thrombus in high resolution; (e, f)
bremsstrahlung single-photon emission computed tomography with integrated CT (SPECT/CT) in
trans-axial and coronal planes show faint, indistinct activity in the same region in low resolution.

Quantification of 90Y activity by PET obtains 13.3.2 TECHNICAL CHALLENGES


reasonably accurate tissue absorbed doses, which OF 90Y PET
may be used to guide postradioembolization man-
agement (Willowson et al., 2015). 90Y PET voxel Clinical 90Y PET is currently in the early phases
dosimetry can also generate dose–volume histo- of development with many technical challenges
grams to graphically describe the heterogeneous to address. Most TOF PET scanners contain
nature of microsphere biodistribution, which can- lutetium-176 within its crystal array that is naturally
not be accounted for using mean absorbed doses radioactive. Coupled with a scant number of posi-
alone (Kao et al., 2013b). trons from 90Y decay, the reconstructed images are
266 Diagnostic reporting using postradioembolization imaging

Table 13.1 Recommendations for diagnostic reporting of postradioembolization 90Y PET/CTa


Continuity of care Postradioembolization 90Y PET/CT is best reported by the same attending
nuclear medicine physician who has followed through the entire planning-
therapy continuum from exploratory angiography and predictive
dosimetry, to 90Y radioembolization.
PET display threshold For detection of nontarget 90Y activity, the operator should actively adjust
setting for nontarget the upper PET visual display threshold setting to deliberately increase the
90Y activity detection background noise to moderate levels.
Criteria for technical 1. 90Y activity present in the majority of targeted tumors, or good overall
success activity coverage of large targeted tumors; and
2. The absence of clinically significant nontarget 90Y activity; and
3. All findings are in keeping with pretherapy radiation planning
expectations.
Criteria for a 1. The complete or near-complete absence of 90Y activity in the majority of
technically targeted tumors, or poor overall activity coverage of large targeted
unsuccessful 90Y tumors; or
radioembolization 2. The presence of any nontarget 90Y activity where 90Y PET dose
quantification predicts a high likelihood of clinically significant radiation
toxicity; or
3. Any other situation where the 90Y activity biodistribution is adversely
inconsistent with pretherapy radiation planning expectations.
Criteria for nontarget 1. Nonrandom pattern of activity distribution; and
90Y activity 2. Conforms morphologically to an untargeted anatomical structure on CT;
with or without
3. A plausible vascular etiology to account for its presence.
Criteria for noise 1. Small, discrete, ovoid activity foci; and
(a) (b)
spikes 2. Random pattern of distribution, which does not conform to underlying
anatomy on CT; and
3. No plausible vascular etiology to account for its presence.
Source: With kind permission from Springer Science+Business Media: EJNMMI Res, Post-radioembolization
yttrium-90 PET/CT—Part 1: Diagnostic reporting, 3, (2013a), 56, Kao, Y.H. et al.
Note: 90 Y PET CT, yttrium-90 positron emission tomography with integrated computed tomography.
a Reproduced under the terms of the Creative Commons Attribution License for an Open Access article (Kao et al.,

2013a).

(a) (b) (c)

(c)
Figure 13.2 Nontarget activity along the falciform ligament to the umbilicus detected on bremsstrah-
lung SPECT/CT (arrows) depicted in (a) coronal, (b) sagittal, and (c) trans-axial planes. The patient
underwent resin microsphere radioembolization of colorectal liver metastases, with bevacizumab 6
weeks prior. Widespread mild nontarget activity was detected throughout the celiac axis, presumed
to be due to stasis and reflux. The patient experienced significant abdominal pain during and after
radioembolization that was managed well with analgesia. 90Y PET was not available. There were no
clinical signs of radiomicrosphere dermatitis on follow-up over 2 months.
13.3 90 Y PET with integrated CT / 13.3.2 Technical challenges of 90Y PET 267

(a) (a) (b)

(b)(a) (b)

(c) Figure 13.4 Trans-axial slice of 90Y PET/CT (a)


and PET (b) inferior to the level of the liver of a
technically successful radioembolization. All of
the apparent activity seen here is noise. Noise
is typically discrete foci of variable intensity in a
random distribution that does not correspond
morphologically to any underlying anatomy
and appears in locations that do not have any
plausible vascular etiology.

radioconcentration, the better the image quality


and vice versa. Noise is worst if the entire liver was
treated with a low total activity of 90Y microspheres
Figure 13.3 Multifocal hepatocellular carcinoma resulting in very low 90Y radioconcentration within
with multiple small tumors. (a) Catheter-directed
the field-of-view. The resultant noisy images may
CT of the proper hepatic artery demonstrates
the hypervascularity of the multiple small tumors. adversely affect the diagnostic accuracy of target
Small tumors in the right lobe are apparent, and nontarget activity assessment.
several of which are subcentimeter in diam- Similarly, the quantitative accuracy of 90Y PET
eter. (b) 90Y PET/CT depicts, in high resolution, in areas of low radioconcentration such as the
discrete focal activity within the small tumors. (c) nontumorous liver and lung are also vulnerable to
Bremsstrahlung SPECT/CT shows focal activity noise. Another problem with lung activity quanti-
only in the two largest tumors; activities in the fication by 90Y PET is the “spill-in” of activity from
other small tumors were indistinguishable from the liver dome into lung bases due to tidal breath-
nontumorous liver activity.
ing (Figure 13.5). This will result in overestimation
of absorbed doses at the lung bases (Figure 13.6)
vulnerable to high background noise (Figure 13.4). and underestimation of the liver dose. Respiratory-
As can be expected, the severity of background gated 90Y PET/CT may be a promising solution and
noise is correlated to the 90Y radioconcentration further research is warranted (Mamawan et al.,
within the field-of-view, that is, the higher the 90Y 2013).
268 Diagnostic reporting using postradioembolization imaging

(a) (b) (c)

Figure 13.5 Nonrespiratory-gated 90Y PET/CT demonstrating misregistration at the right diaphragm.
Horizontal red lines depict the extent of misregistration between PET and CT components. Parts a, b,
and c are coronal 90Y PET, coronal CT, and coronal fused 90Y PET/CT images, respectively.

(a) (b)

(c)

Figure 13.6 Activity “spill over” from the liver dome into the right lung base (arrows) depicted by vol-
umetric isocontour thresholding of 90Y PET/CT. This may affect the accuracy of activity quantification
at the right lung base. Parts a, b, and c are axial, coronal, and sagittal reconstructions, respectively.

The benefits and challenges associated with is considerably noisier than conventional PET trac-
quantitative 90Y PET/CT imaging are described in ers and may seem uninterpretable. Fortunately, the
additional detail in Chapter 11. problem of noise may be ameliorated using some
simple qualitative techniques to facilitate diagnos-
tic reporting.
13.4 PrINCIPLES OF Recommendations on the general technique
DIaGNOStIC rEPOrtING for diagnostic reporting of postradioemboliza-
tion 90Y PET are summarized in Table 13.1 (Kao
90Y PET is vulnerable to noise due to the low–true et al., 2013a). Many of its recommendations are
coincidence rate and natural radioactivity from also applicable for bremsstrahlung SPECT/CT, but
lutetium-based PET crystals. At the outset, the within its inherent limitations of poorer spatial
visual quality of the reconstructed 90Y PET images resolution and lack of quantitative accuracy.
13.4 Principles of diagnostic reporting / 13.4.2 Target activity 269

13.4.1 CONTINUITY OF CARE is within pretherapy planning expectations. The


first step is to manually adjust the PET upper dis-
Radioembolization is a multistage continuum play threshold to qualitatively suppress the visual
requiring close interdisciplinary coordination and appearance of noise (Figure 13.7). This is because
communication. Case-specific technical complexi- target activity is usually more intense than nontar-
ties often influence the final angiographic approach get activity and background noise. It is important
and radiation plan. Each patient’s planning-therapy to note that normal microsphere biodistribution is
continuum is unique and continuity of care is always heterogeneous at both the microscopic and
paramount for clinically meaningful diagnos- macroscopic levels. Therefore, “clumps” of activity
tic reporting of postradioembolization imaging. in tumor, nontumorous liver, or lungs should not be
Therefore, it is preferred that the same members routinely disregarded as noise.
of the multidisciplinary team follow through the
entire workflow from exploratory angiography to
radioembolization. If any team member is different (a) (b)
between the planning and treatment stages, hando-
ver may risk inadvertent omission of crucial tech-
nical details or their significance may not be fully
appreciated by the new member.
For diagnostic reporting of postradioemboliza-
tion imaging, the two key questions are whether
the radioembolization was technically successful (c) (d)
and whether any clinically significant nontarget
activity was detected. To answer these questions
meaningfully, the reporting doctor should have
case-specific knowledge of the target arterial ter-
ritories (e.g., whole-liver, lobar, segmental, subseg-
mental), treatment intent (e.g., Palliative, lobectomy,
segmentectomy, sequential, intentional sparing),
(e) (f )
arteries-at-risk, and any last minute deviations from
the intended angiographic plan due to unforeseen
on-table events. In general, 90Y activity biodistribu-
tion that is within pretherapy planning expectations
may be considered as a technical success. It should
be reiterated that “technical success” is a qualitative
term that has little or no bearing on “clinical success”
unless tissue absorbed doses are known by predictive (g) (h)
dosimetry or 90Y PET quantification.
Even prior to looking at the postradioemboliza-
tion images, the reporting doctor should already
have an expectation of what the 90Y activity biodistri-
bution should be, as was simulated by Tc-99m MAA
SPECT/CT. If the Tc-99m MAA biodistribution
appears unexpectedly discordant to the 90Y activity Figure 13.7 The importance of qualitative adjust-
biodistribution, the reporting doctor should attempt ment of the PET upper display threshold to mini-
to explain this based on the case-specific knowledge mize the visual appearance of noise for target
to further enhance patient management. activity assessment. This series of images depicts
the same trans-axial slice of a 90Y PET/CT over
four different PET upper display thresholds: (a, b)
13.4.2 TARGET ACTIVITY 0% (40 kBq/mL); (c, d) 2% (190 kBq/mL); (e, f)
20% (2,030 kBq/mL); and (g, h) 100% (10,430
The aim of target activity assessment is to determine kBq/mL). The left lobe tumor with heterogeneous
whether the biodistribution of target 90Y activity 90Y activity is best seen in (g, h).
270 Diagnostic reporting using postradioembolization imaging

For hypervascular tumors, 90Y activity is nor- tumors such as those partially treated by other
mally more intense at its hypervascular periphery modalities usually appear relatively photopenic as
becoming less intense toward its center. For large compared with its surrounding nontumorous liver
or massive tumors, the heterogeneous micro- parenchyma (Figure 13.9).
sphere biodistribution is often visually detectable It may be possible to comment on 90Y activity
as heterogeneous clumps of activity. In these cases, within subcentimeter tumors if the focally implanted
technical success requires good circumferential radioconcentration is high. Within the limitations of
coverage of 90Y activity throughout the tumor 90Y PET spatial resolution, it is sometimes necessary

with minimal gaps of absent activity. It is a nor- to comment on the presence or absence of activity
mal finding in massive tumors for the intensity of within small but critical target lesions such as portal
90Y activity to gradually decrease toward its core vein tumor thrombosis. For example, the absence of
due to reduced arterial penetration and central significant 90Y activity within targeted portal vein
necrosis (Figure 13.8). From a clinical perspective, tumor thrombosis is ominous for early progressive
this means that a complete response is difficult to disease and warrants vigilant follow-up or adjuvant
achieve for massive tumors using radioemboliza- treatment modalities (Figure 13.10).
tion alone because the total prescribed 90Y activ-
ity would have been dosimetrically constrained for
safety to the nontumorous liver or lungs. 13.4.3 NONTARGET ACTIVITY
For multiple small- to medium-sized tumors, VERSUS NOISE
technical success requires 90Y activity to be detected
in the majority of targeted tumors, within the limi- Assessment of nontarget activity by 90Y PET is
tations of 90Y PET spatial resolution. Hypovascular challenging due to background noise, which may
confuse the reporting doctor unless an appropriate
(a) diagnostic technique is applied. Until the arrival
of better imaging and reconstruction protocols for
90Y PET to minimize noise, the following simple

qualitative techniques may improve the accuracy


of nontarget activity detection.
First, the reporting doctor should be aware that
noise spikes might be of greater visual intensity
than nontarget activity. Noise typically appears as
ovoid foci that are randomly distributed through-
out the field-of-view and do not correlate with any
(b)
angiographically plausible anatomical structure
(Figure 13.4). The visual intensity of nontarget
activity may be subtle or mild because it reflects
the amount of implanted 90Y microspheres. This
means that the visual appearance of nontarget
activity may vary widely; hence any suspicious
activity should not be disregarded as noise based
on visual intensity alone.
Nontarget activity should also be interpreted
in conjunction with the reporting doctor’s
impression of the likelihood of nontarget tis-
sue toxicity. The absence of clinical symptoms at
Figure 13.8 Massive hepatocellular carcinoma
the time of postradioembolization imaging does
in the right lobe with central necrosis. (a) 90Y
PET/CT depicts in high resolution the inher-
not exclude a qualitative diagnosis of nontarget
ently heterogeneous tumor activity biodistribu- activity. This is because clinical symptoms may
tion. Photopenic regions correspond to central not manifest until days, weeks, or months later
necrosis. (b) CT liver in the portovenous phase depending on the organ involved and the nontar-
for correlation. get absorbed dose.
13.4 Principles of diagnostic reporting / 13.4.3 NONTARGET ACTIVITY VERSUS NOISE 271

(a) (b)

(c) (d)

Figure 13.9 Hypovascular intrahepatic cholangiocarcinoma in the right lobe. (a) Catheter-directed
CT of the proper hepatic artery shows that the targeted tumor is predominantly hypovascular. (b)
18-Fluorodeoxyglucose (FDG) PET/CT shows that the tumor is mostly viable with only a small amount
of central necrosis. (c, d) 90Y PET/CT after radiomicrosphere segmentectomy depicts in high resolu-
tion the heterogeneous activity biodistribution at the tumor periphery with low activity within the
tumor mass, consistent with a hypovascular tumor.

As nontarget activity may be of lower visual anatomical structure is unlikely to represent non-
intensity than background noise, the reporting target activity and is probably noise. The absence
doctor should first qualitatively adjust the PET dis- of a proven artery-at-risk by retrospective review of
play threshold to deliberately increase background angiography does not exclude a diagnosis of non-
noise to moderate levels. This counterintuitive tech- target activity because the culprit artery may not
nique facilitates the visual detection of any nonran- always be identified on angiography.
dom pattern among a random noise background. Nontarget activity in the stomach, duodenum,
The rotating maximum intensity projection (MIP) and gallbladder should appear in a linear pattern
is useful to detect any nonrandom activity pattern conforming to its walls (Figure 13.11). In untargeted
protruding against target tissue activity. Any non- liver, nontarget activity should conform morpho-
random activity pattern detected on the rotating logically to the parenchyma of untargeted lobe or
MIP should be pursued on the PET/CT (or PET/ segments (Figure 13.12). Suspicious but visually
MR) images in axial, coronal, and sagittal planes. subtle activity is diagnostically challenging and
The CT or MR images should be carefully exam- often indeterminate for nontarget activity versus
ined for any corresponding anatomical structures noise. Fortunately, such indeterminate cases are
conforming morphologically to the visual distribu- usually clinically insignificant because the nontarget
tion of the suspected nontarget activity. This should absorbed dose is likely to be low.
also be supported by an angiographically plausible Both 90Y PET and bremsstrahlung SPECT/CT are
theory to explain the presence of nontarget activity. usually not respiratory-gated and therefore vulner-
The converse is true: any activity pattern that does able to misregistration. This problem is worst when
not correspond to an angiographically plausible assessing for nontarget activity in viscera that lie
272 Diagnostic reporting using postradioembolization imaging

(a) (b)

(c) (d)

(e) (f )

(g) (h)

Figure 13.10 Multifocal hepatocellular carcinoma with portal vein tumor thrombosis. (a, b) Portal vein
tumor thrombosis (arrows) depicted on portal venous phase of triphasic CT liver in trans-axial and cor-
onal planes. (c, d) 90Y PET/CT shows, in high resolution, subtle 90Y activity within the portal vein tumor
thrombus, which is unlikely to be effective. (e, f) Bremsstrahlung SPECT/CT was indeterminate for the
presence or absence of focal activity within the portal vein tumor thrombus due to visual interference
from adjacent liver activity. (g, h) Follow-up triphasic CT liver 3 months after radioembolization shows
progression of the portal vein tumor thrombosis, depicted here in the portal venous phase in trans-
axial and coronal planes. This clinically validates the 90Y PET/CT finding of subtle, ineffective activity
within the portal vein tumor thrombosis.

closely adjacent to the liver serosa such as the gastric 13.4.4 TECHNICAL FAILURE
pylorus, proximal duodenum, or gallbladder fundus.
Due to the poor spatial resolution of bremsstrahlung The fundamental premise of radioembolization is
SPECT/CT, these areas are often indeterminate for to balance safety and efficacy within a multidisci-
the nontarget activity. The improved spatial resolu- plinary framework. Technical failure has occurred
tion of 90Y PET partially ameliorates this problem when the 90Y activity biodistribution is adversely
with additional quantitative capability. inconsistent with pretherapy planning expectations.
13.4 Principles of diagnostic reporting / 13.4.4 Technical failure 273

(a) (b)

Figure 13.11 Nontarget 90Y activity in the proximal duodenum. 90Y PET/CT shows mild nontarget
activity in a linear morphology corresponding to the proximal duodenal wall. Nontarget activity can
be appreciated both on the fused 90Y PET/CT (a) and 90Y PET image (b). 90Y PET quantification was not
performed. The patient was clinically asymptomatic on follow-up.

(a) (b)

(c) (d)

Figure 13.12 Nontarget 90Y activity in the untargeted left lobe. (a) Large hypovascular hepatocel-
lular carcinoma of the right lobe depicted by catheter-directed CT of the right hepatic artery. (b)
Bremsstrahlung SPECT/CT shows, in low resolution, subtle diffuse bremsstrahlung activity in the
untargeted left lobe (arrows). (c, d) 90Y PET/CT shows, in high resolution, nontarget activity in a non-
random distribution conforming to the anatomy of the untargeted left lobe. The nontarget activity
was probably due to mild microsphere reflux, arterioportal shunting, or both.

In other words, significant irreversible technical failure should prompt an urgent clinical review for
complications had occurred during radioemboli- early management in terms of adjuvant treatment or
zation with the potential to cause severe radiomi- mitigative action to minimize potential toxicity.
crosphere toxicity. Technical failure is, therefore, a The specific definition of technical failure dif-
serious diagnosis that should not be made without fers depending on the size and distribution of the
due consideration because it implies a high likeli- targeted tumors and the overall treatment intent.
hood of adverse clinical outcomes and also nega- For tumors, technical failure generally means very
tively impacts team morale. A diagnosis of technical poor tumor 90Y activity coverage and early disease
274 Diagnostic reporting using postradioembolization imaging

progression is expected. For massive tumors, tech- should be performed because visual assessment
nical failure means that large regions of the tumor alone is subjective. The nontarget tissue absorbed
are devoid of 90Y activity, excluding central necrosis. dose provides an objective and radiobiologically
For nontarget activity, technical failure means rational basis to predict toxicity, which in turn
that a significant amount of 90Y activity has been impacts mitigative action. Nontarget activity may
detected within nontarget tissue and severe toxicity be assumed to be clinically insignificant only if
is likely. However, visual assessment alone is subjec- visually subtle; all other cases should be objectively
tive and unreliable for toxicity prediction. Therefore, supported by absorbed dose quantification.
any nontarget activity of clinical concern should be The likelihood and severity of nontarget toxicity
guided by 90Y PET absorbed dose quantification. depends on the organ involved and absorbed dose
biodistribution and should always be assessed on a
13.4.5 QUANTIFICATION OF case-specific basis. Radiomicrosphere dose–response
NONTARGET ABSORBED data for nontarget tissue toxicity are currently scarce.
DOSE To fill this knowledge gap, dose–response experi-
ences of external beam radiotherapy may serve as
For clinically meaningful diagnostic reporting, an interim guide using mathematical extrapolations
nontarget activity should be reported together such as the biologically effective dose (Cremonesi et
with its likelihood of toxicity. To achieve this, 90Y al., 2014). However, such extrapolations must be cau-
PET quantification of the nontarget absorbed dose tiously used because the radiobiology of radioactive

(a)
6PT CT 3D

6
Avg: 1076665 Bq/mL, 19 HU
Max: 3424908 Bq/mL, 100 HU
Volume: 22.47 cm3
Threshold: 499018 Bq/mL

(b) (c)

6PT CT 3D 6PT CT 3D

6 6
Avg: 1076665 Bq/mL, 19 HU Avg: 1076665 Bq/mL, 19 HU
Max: 3424908 Bq/mL, 100 HU Max: 3424908 Bq/mL, 100 HU
Volume: 22.47 cm3 Volume: 22.47 cm3
Threshold: 499018 Bq/mL Threshold: 499018 Bq/mL

Figure 13.13 Absorbed dose quantification of nontarget activity in the gastric pylorus by 90Y PET.
A volume-of-interest was defined in the pylorus by volumetric isocontour thresholding and its mean
radioconcentration obtained. After decay correction to the time of radioembolization, a mean
absorbed dose of approximately 65 Gy was obtained. This patient developed chronic abdominal
pain and pyloric ulceration seen on endoscopy 3 months later. Parts a, b, and c are axial, coronal, and
sagittal reconstructions, respectively.
13.4 Principles of diagnostic reporting / 13.4.6 Verification of absorbed doses 275

microspheres is different to that of external beam to the current paucity of data, further research into
radiotherapy and also between different types of nontarget dose–response is warranted to guide
radioactive microspheres. toxicity prognostication.
90Y PET absorbed dose quantification in hol-

low viscus is challenging. A simple solution may 13.4.6 VERIFICATION OF ABSORBED


be to define a volume-of-interest to obtain its mean DOSES
radioconcentration. This is then decay corrected
and the 90Y absorbed dose coefficient (approxi- Modern personalized radioembolization utilizes
mately 50 Gy per GBq/kg) applied to obtain its patient-specific tomographic parameters to opti-
mean absorbed dose (Figure 13.13). Using this mize the brachytherapy radiation plan. If a scien-
method, preliminary data for 90Y resin micro- tifically sound and meticulous method of pretherapy
spheres found that approximately 49 Gy to a local- radiation planning had been used for 90Y activity
ized area of the gastric wall may cause gastritis, prescription (e.g., “artery-specific SPECT/CT parti-
65 Gy may result in ulceration, whereas less than tion model”; Kao et al., 2012), then technical suc-
18 Gy may be asymptomatic; 53 Gy to the duode- cess means that the intended radiation plan may
num may cause duodenitis (Kao et al., 2013b). Due be assumed to be valid within the general limits of

(a) (b)

(c) (d)

Figure 13.14 Absorbed dose quantification of a portal vein tumor thrombus by 90Y PET. (a) Triphasic
CT liver in the arterial phase demonstrates a large contrast-enhancing portal vein tumor throm-
bus (arrow). (b, c) A volume-of-interest approximating the activity boundaries of the portal vein
tumor thrombus was defined by volumetric isocontour thresholding and its mean radioconcentra-
tion obtained. After decay correction to the time of radioembolization, a mean absorbed dose of
approximately 248 Gy was obtained. (d) Follow-up triphasic CT liver in the arterial phase at 4 months
postradioembolization shows a slight decrease in lesion size and a complete lack of contrast enhance-
ment within the portal vein tumor thrombus (arrow), suggesting a complete response; this clinically
validates the mean radiation absorbed dose quantified by 90Y PET.
276 Diagnostic reporting using postradioembolization imaging

dosimetric uncertainty (Kao et al., 2013c; Song et desired, for example, portal vein tumor thrombus
al., 2015). This means that the treatment response (Figure 13.14).
is expected to be in accordance to the prescribed One method of 90Y PET quantification is to
tissue absorbed doses. In such cases, postradio- apply the “local deposition method” (Chapter
embolization verification of absorbed doses by 90Y 12) to obtain the mean absorbed dose and dose–
PET quantification is unnecessary unless for qual- volume histogram within a volume-of-interest
ity control, research, or dose–volume histograms, (Figure 13.15) (Kao et al., 2013b; Pasciak et al.,
or if the absorbed dose of a specific lesion is 2014). Using this analysis, predictive dosimetry

(a) (b) (c)

Length: 3.974 cm (61.652 pts)

Length: 3.458 cm (63.652 pts)

(d) (e)

Length: 2.116 cm (30.849 pts)

Length: 1.822 cm (26.550 pts)

(f ) 100 Volume 74.4 cm3


90 T/N ratio 11.8
80 Mean 425.3 Gy
Tumor volume (%)

70 Minimum 30.1 Gy
60 Maximum 1618.9 Gy
50
40
30
20
10
0
0 100 200 300 400 500 600 700 800 900 1000 110012001300140015001600
Tumor radiation absorbed dose (Gy)

Figure 13.15 90Y PET tumor voxel dosimetry and dose–volume histogram using the local deposi-
tion method. (a) Triphasic CT liver in the arterial phase shows a right lobe hepatocellular carcinoma
measuring 4.0 × 3.5 cm. (b) 90Y PET/CT depicts activity biodistribution in high resolution, with intense
tumor activity and low-grade activity in nontumorous liver. (c) Corresponding trans-axial slice of the
90Y PET display used for manual contouring of tumor volume-of-interest for voxel dosimetry, indicated

by small black dots. (d) Isodose map of the corresponding trans-axial slice of the right liver lobe pro-
vides a visual representation of dose heterogeneity within the target arterial territory and displays the
full range of delivered dose from 0 Gy to >1600 Gy. (e) Follow-up triphasic CT liver in arterial phase
5.5 months later shows a noncontrast-enhancing hypodensity with significant size reduction to 2.1
× 1.8 cm, representing a complete response. (f) Dose–volume histogram generated by 90Y PET voxel
dosimetry from the tumor volume-of-interest shown in (c). Mean, minimum, and maximum tumor
absorbed doses were 425 Gy, 30 Gy, and 1619 Gy, respectively; D70 was >210 Gy, where D70 is the
minimum absorbed dose to 70% tumor volume.
13.5 Economics of 90Y PET 277

based on Tc-99m MAA SPECT/CT was shown to


be accurate for tumor absorbed doses with a low 13.5 ECONOMICS OF 90Y PEt
mean bias of +6.0% (95% confidence interval –1.2%
to +13.2%) in a subset of highly select tumors (Kao A practical challenge of 90Y PET is its relatively
et al., 2013b). long acquisition time as compared with conven-
Tissue mean absorbed doses may also be calcu- tional PET tracers such as 18-fluorodeoxyglucose
lated using simple count ratios to obtain the “true” (FDG). Due to a very low 90Y positron fraction, a
tumor-to-normal liver (T/N) ratio, analogous to longer acquisition per bed is preferred. Today’s
that estimated by Tc-99m MAA during prether- TOF scanners typically acquire 90Y PET at 15–20
apy planning. Input of the true T/N ratio back minutes per bed position. If the liver is markedly
into tissue masses and lung shunt fraction as per enlarged, two bed positions may be required,
Medical Internal Radiation Dose (MIRD) macro- increasing the total PET acquisition time to
dosimetry (i.e., “Partition Model”) (Ho et al., 1996) 30–40 minutes. In comparison, a whole-body
will obtain more accurate tissue mean absorbed FDG PET/CT by a TOF scanner takes 20–30 min-
doses. Using this analysis, good correlations were utes to complete. Hence, the economic impact of
found for mean absorbed doses by Tc-99m MAA performing such long 90Y PET scans for a single
compared with 90Y PET for both tumor (r = 0.64; patient cannot be ignored, especially in high-
p < .01) and nontumorous liver (r = 0.71; p < .001) throughput PET centers.
(Song et al., 2015). A total scan time of 40 minutes for two-bed
Semiempirical 90Y activity prescription such as positions is probably at the limit of tolerance for
the “body surface area method” for resin micro- most patients. Any further increase in acquisi-
spheres has no radiobiologically rational basis to tion time may risk patient discomfort, movement,
establish any dose–response relationships, unless and misregistration (Figure 13.16). Unless future
90Y PET quantification is retrospectively performed.
research can significantly shorten the PET acqui-
As an inherent conceptual limitation of the semiem- sition time without compromising image quality,
pirical paradigm, patient-specific tissue absorbed extending the PET field-of-view from the abdo-
doses are unknown at the time of 90Y activity pre- men into the lungs will result in an impractically
scription. At the discretion of the treating team, long total scan time of 45–80 minutes over three
90Y PET quantification may be retrospectively per-
to four bed positions. Any respiratory gating will
formed to discover what the tissue absorbed doses compound the total scan time even longer.
actually were, albeit too late for any absorbed dose If 90Y PET of both the lung and liver is clini-
modification. This situation is similar for glass cally indicated, for example, patients with high
microspheres, where 90Y activity prescription is
generally based on a mean absorbed dose averaged
across the entire target arterial territory.
For the lung, absorbed dose verification by 90Y
PET is more technically challenging. First, tidal
breathing may overestimate the lung absorbed
dose at the lung bases due to “spill in” of activ-
ity from the liver dome (Figures 13.5 and 13.6).
Respiratory gating may be a possible solution
(Mamawan et al., 2013). Second, the lung radio-
concentration within the PET field-of-view may
be too low for accurate quantification. However, it
14.9 mm (2D)
may be theoretically possible to indirectly calculate 14.4 mm (2D)
the total lung activity as the difference between 12.4 mm (2D)
the total injected activity and whole-liver activity
quantified by 90Y PET. The lung mean absorbed Figure 13.16 Trans-axial PET/CT misregistra-
dose may then be calculated using the patient- tion of up to 1.5 cm in the liver due to patient
specific lung mass estimated by CT densitovolum- movement during a 15 minutes per bed 90Y PET
etry (Kao et al., 2014b). acquisition.
278 Diagnostic reporting using postradioembolization imaging

lung shunting where knowledge of the true lung REFERENCES


absorbed dose is clinically relevant, a possible solu-
tion may be to break up the lung and liver into two Cremonesi, M. et al. (2014). Radioembolization of
separate acquisitions to allow the patient to rest hepatic lesions from a radiobiology and dosi-
between the two scans. All 90Y PET quantifica- metric perspective. Front Oncol 2014;4:210.
tion must be decay corrected to the time of radio- Ho, S. et al. (1996). Partition model for estimating
embolization. The overall economic feasibility of radiation doses from yttrium-90 microspheres
90Y PET will vary depending on the healthcare in treating hepatic tumours. Eur J Nucl Med
financial model and research grant availability of 23:947–952.
each country and institution. Jiang, M. et al. (2012). Segmental perfusion dif-
ferences on paired Tc-99m macroaggregated
albumin (MAA) hepatic perfusion imaging
13.6 CONCLUSIONS and yttrium-90 (90Y) bremsstrahlung imag-
ing studies in SIR-Sphere radioembolization:
Associations with angiography. J Nucl Med
90Y PET is technically superior to bremsstrahlung
Radiat Ther 3:122.
SPECT/CT and should be preferred where avail-
Kao, Y.H. et al. (2011). Imaging discordance
able. Continuity of care is central to clinically
between hepatic angiography versus Tc-99m-
meaningful diagnostic reporting of postradioem-
MAA SPECT/CT: A case series, technical
bolization imaging. Qualitative and quantitative
90Y PET are interrelated and inseparable compo-
discussion and clinical implications. Ann Nucl
Med 25:669–676.
nents that should be interpreted in the context of
Kao, Y.H. et al. (2012). Image-guided personal-
each other. Further research on 90Y PET is required
ized predictive dosimetry by artery-specific
to improve the quality of reconstructed images and
SPECT/CT partition modeling for safe and
accuracy of absorbed dose quantification and to
effective 90Y radioembolization. J Nucl Med
investigate dose–response relationships in micro-
53:559–566.
sphere radiobiology.
Kao, Y.H. et al. (2013a). Post-radioembolization
yttrium-90 PET/CT—Part 1: Diagnostic report-
ing. EJNMMI Res 3:56.
ACKNOWLEDGMENTS Kao, Y.H. et al. (2013b). Post-radioembolization
yttrium-90 PET/CT—Part 2: Dose–response
The following members and their institutions are and tumor predictive dosimetry for resin
acknowledged for their contributions: Anthony microspheres. EJNMMI Res 3:57.
Goh and David Ng, Department of Nuclear Kao, Y.H. (2013c). A clinical dosimetric perspec-
Medicine and PET, Singapore General Hospital, tive uncovers new evidence and offers new
Singapore; Jeffrey Steinberg, Jianhua Yan, and insight in favor of 99mTc-macroaggregated
David Townsend, Agency for Science Technology albumin for predictive dosimetry in 90Y resin
and Research—National University of Singapore microsphere radioembolization. J Nucl Med
Clinical Imaging Research Centre, Singapore; 54:2191–2192.
Mark Goodwin, Sze Ting Lee, and Andrew Kao, Y.H. et al. (2014a). Non-target activ-
Scott, Department of Radiology, Department ity detection by post-radioembolization
of Molecular Imaging and Therapy, Austin yttrium-90 PET/CT: Image assessment
Health, Melbourne, Australia; Meir Lichtenstein, technique and case examples. Front
Department of Nuclear Medicine, The Royal Oncol 4:11.
Melbourne Hospital, Melbourne, Australia; and Kao, Y.H. et al. (2014b). Personalized predic-
Richard Dowling, Department of Radiology, The tive lung dosimetry by technetium-99m
Royal Melbourne Hospital, Melbourne, Australia; macroaggregated albumin SPECT/CT for
Jan Boucek, Department of Nuclear Medicine, Sir yttrium-90 radioembolization. EJNMMI Res
Charles Gairdner Hospital, Perth, Australia. 4:33.
References 279

Lhommel, R. et al. (2009). Yttrium-90 TOF PET Song, Y.S. et al. (2015). PET/CT-based dosimetry
scan demonstrates high-resolution biodistri- in 90Y-microsphere selective internal radia-
bution after liver SIRT. Eur J Nucl Med Mol tion therapy: Single cohort comparison with
Imaging 36:1696. pretreatment planning on 99mTc-MAA imag-
Mamawan, M.D., Ong, S.C., Senupe, J.M. (2013). ing and correlation with treatment efficacy.
Post-90Y radioembolization PET/CT scan with Medicine (Baltimore) 94:e945.
respiratory gating using time-of-flight recon- Walrand S et al. (2011). Yttrium-90-labeled
struction. J Nucl Med Technol 41:42. microsphere tracking during liver selective
Nickles, R.J. et al. (2004). Assaying internal radiotherapy by bremsstrahlung
and PET imaging of yttrium-90: 1 pinhole SPECT: Feasibility study and evalua-
>>34 ppm > 0. IEEE Nuclear Science tion in an abdominal phantom. EJNMMI Res
Symposium Conference Record, October 1:32.
16–22, 2004; Casaccia, Italy. New York, NY: Willowson, K.P., Tapner, M.; QUEST Investigator
IEEE; 2004:3412–3414. Team, Bailey, D.L. (2015). A multicentre
Pasciak, A.S., Bourgeois, A.C., Bradley, Y.C. comparison of quantitative (90)Y PET/CT for
(2014). A comparison of techniques for (90) dosimetric purposes after radioemboliza-
Y PET/CT image-based dosimetry following tion with resin microspheres: The QUEST
radioembolization with resin microspheres. Phantom Study. Eur J Nucl Med Mol Imaging
Front Oncol 4:121. 42:1202–1222.
Salem, R. et al. (2011). Research report- Wissmeyer, M. et al. (2011). 90Y Time-of-flight
ing standards for radioembolization of PET/MR on a hybrid scanner following liver
hepatic malignancies. J Vasc Interv Radiol radioembolisation (SIRT). Eur J Nucl Med Mol
22:265–278. Imaging 38:1744–1745.
14
The use of postprocedural imaging in
the medical management of patients

AUSTIN C. BOURGEOIS, MARCELO S. GUIMARAES, YONG C. BRADLEY,


CHRISTOPHER HANNEGAN, AND ALEXANDER S. PASCIAK

14.1 Introduction 281 14.3.4 Gastrointestinal nontarget


14.1.1 Chapter overview 281 embolization 295
14.1.2 Background 282 14.3.5 Radiation pneumonitis and
14.2 Postprocedural imaging: findings and other sites of nontarget
significance 282 embolization 296
14.2.1 Imaging surveillance protocols 282 14.4 Posttreatment 90Y imaging 296
14.2.2 Preprocedural imaging with 14.4.1 Evaluating nontarget
prognostic significance 283 embolization with
14.2.3 Normal response to therapy 284 posttreatment 90Y imaging 296
14.2.4 Follow-up imaging assessment 14.4.2 Prognostication with 90Y PET/CT 297
criteria in predicting treatment 14.5 Treatment modification with 90Y PET/CT 299
response 287 14.5.1 Interprocedural treatment
14.3 Imaging of complications 291 modification 299
14.3.1 Hepatic toxicity 291 14.5.2 Interprocedural treatment
14.3.2 Biliary effects 292 modification 299
14.3.3 Hepatic abscess 295 14.6 Conclusions 299
References 299

risks to surgical interventional and other forms


14.1 INtrODUCtION of liver-directed therapy (i.e., fulminant hepatic
failure), it can result in a variable constellation
14.1.1 CHAPTER OVERVIEW of both normal treatment effects and possible
complications. Thus, image interpretation fol-
Hepatic radioembolization with yttrium-90 (90Y) lowing 90Y can be confusing, particularly when
microspheres is unique in many ways compared performed in a vacuum of clinical information
with other methods of treating hepatic malig- and without a robust understanding of radio-
nancy, including the use of radiation, size of the embolization. This chapter provides a detailed
infused microsphere, the number of particles discussion of the imaging and correlative clini-
administered, and variable technical consider- cal findings following 90Y therapy with an
ations involved in the infusion. While radioem- emphasis on clinical relevance and underlying
bolization imparts some of the same procedural mechanisms.

281
282 The use of postprocedural imaging in the medical management of patients

It is important to consider interventional radi- the increased arterial perfusion to tumors relative
ologists, radiation oncologists, nuclear medi- to that of normal liver, that ranges from approxi-
cine radiologists, and medical physicists may be mately 3:1 in colorectal metastasis to often greater
involved in a 90Y treatment planning and deliv- than 5:1 for HCC, as described in Chapter 5.
ery. Each possesses some scope of formal train- Whereas TACE derives a significant component
ing regarding imaging physics and interpretation. of its efficacy from small vessel occlusion leading
Because those involved in the care of 90Y patients to tumor ischemia, 90Y provides a localized radia-
understand imaging, the integration of advanced tion therapy with a less significant embolic effect
imaging modalities into routine practice has (Kennedy et al., 2007). This fundamental differ-
evolved at a rapid pace. For example, a number of ence provides the framework for interpreting the
methods of directly imaging 90Y have been imple- effects and complications of radioembolization,
mented into clinical practice, allowing for early an emphasis of this chapter. Before proceeding, it
evaluation of efficacy and detection of potentially is important to briefly review the basic principles
deleterious nontarget embolization (NTE). These of glass (Therasphere® BTG, Ontario, Canada) or
have been discussed to some degree in Chapter 13. resin (SIR-Spheres®, SIRTex Technology Pty, Lane
This chapter provides a summary of the imaging Cove, Australia) 90Y microspheres, as described in
protocols that are routinely implemented in post- Chapter 1. In particular, the radiation biology of
procedural surveillance and discusses the role of radioembolization (Chapter 8) will be important
direct 90Y imaging in its widely accepted and inves- in understanding the discussion in this chapter,
tigational settings. which will focus in part on adverse effects.
While the principle that radioembolization is
14.1.2 BACKGROUND a form of transarterial brachytherapy contributes
to its efficacy and tolerability, it also directly con-
As discussed in detail in Chapter 2, surgical resec- tributes to its complications and adverse effects.
tion and transplantation remain the mainstay of In particular, NTE to normal liver tissue and
curative therapy for patients with hepatic malig- extrahepatic soft tissues can be clinically signifi-
nancy (Poon et al., 2002). Unfortunately, the vast cant, even when using the most rigorous preven-
majority of patients with both hepatocellular car- tative measures (Riaz, 2014). While Chapter 13
cinoma (HCC) and liver metastasis are not candi- partially focused on identifying NTE using the
dates for surgical cure upon presentation as a result posttreatment 90Y positron emission tomography/
of factors such as poor hepatic reserve, advanced computed tomography (PET/CT), this chapter
tumors, tumor location, and/or presence of extra- describes the role of routine surveillance imag-
hepatic disease (Poon et al., 2002). For patients ing in the continuing care of patients who have
who are not surgical candidates, locoregional ther- received radioembolization.
apies (LRT) such as ablation, transarterial chemo-
embolization (TACE), and radioembolization with
90Y microspheres play an important role in pallia-
14.2 POStPrOCEDUraL
tion and may provide a significant survival bene-
fit (Salem et al., 2002; Higgins and Soulen, 2013;
IMaGING: FINDINGS aND
Salem et al., 2013b; Bargellini, 2014; Bester et al., SIGNIFICaNCE
2014). LRTs may also be coupled with surgery in
order to allow patients to maintain transplant can- 14.2.1 IMAGING SURVEILLANCE
didacy while awaiting a donor liver, to debulk dis- PROTOCOLS
ease, or to downstage marginal surgical candidates
to potentially curative surgical resection (Braat, It cannot be understated that the primary outcome
2014; Khan, 2014). in evaluating a cancer therapy is its impact on sur-
Transarterial LRTs including TACE and radio- vival. Tumor response to a therapy is nonetheless
embolization play a particularly important role in an important surrogate marker of treatment suc-
the treatment of patients with extensive or mul- cess (Singh and Anil, 2013). In the context of 90Y,
tifocal hepatic tumor burden. These treatments follow-up imaging generally consists of contrast-
capitalize on the dual blood supply to the liver, and enhanced CT, magnetic resonance imaging (MRI),
14.2 Postprocedural imaging / 14.2.2 Preprocedural imaging with prognostic significance 283

or PET/CT. Ideally, follow-up examinations are serial examinations performed at 3-month intervals
performed identically to preprocedural imaging thereafter (Salem and Thurston, 2006). This meth-
to allow for direct comparison. In clinical prac- odology was adopted from experience with che-
tice, the preferred imaging modality varies widely moembolization early in the investigation of
based on resource availability, as well as patient radioembolization and remains in widespread use.
and cost considerations (Attenberger et al., 2015). While there is little foundational evidence for this
Contrast-enhanced CT (CECT) plays a primary or many other imaging follow-up regimens after
role in surveillance in many institutions, given 90Y, posttherapy surveillance has been the subject

its widespread availability and relatively low of at least one large study. Boas et al. (2015) exam-
costs (Boas et al., 2015). When compared with ined 1766 patients who underwent locoregional
CT, contrast-enhanced MRI provides improved therapy for treatment of HCC. The authors found
sensitivity for detecting hepatic tumors and may that disease recurrence occurs from 0 to 9 months
detect additional hepatic lesions in as many as 30% after treatment in the vast majority of patients and
of patients (Kim et al., 2015). However, the use peaks at 3 months. Using these data, the authors
of MRI may be limited in patients with implant- suggested that a “front loaded” surveillance proto-
able electronic devices, poor pulmonary function col would provide the appropriate balance of cost
who are unable to comply with required breathing optimization and reduction of diagnostic delay. The
instructions, and those unable to tolerate the rela- recommended protocol includes surveillance CT
tively lengthy MRI acquisition time. or MRI at 2, 4, 6, 8, 11, 14, 18, 24 months after 90Y
2-Deoxy-2-18fluoroglucose PET/CT (18FDG- therapy (Boas et al., 2015). It should be emphasized
PET/CT) may also be especially useful for charac- that any imaging protocols should include a certain
terizing classically hypermetabolic tumors such degree of flexibility and be adjusted according to
uveal melanoma metastasis (Eldredge-Hindy patient-specific preprocedural risk factors, change
et al., 2014). PET imaging provides a quantita- in tumor markers, or clinical evidence of progres-
tive reflection of cellular metabolism by imaging sive disease or complication.
photons emitted from the glucose analog 18FDG
(Soydal et al., 2013). Using PET, important prog- 14.2.2 PREPROCEDURAL IMAGING
nostic information regarding treatment response WITH PROGNOSTIC
to radioembolization can be extrapolated in the SIGNIFICANCE
preprocedural and early postprocedural periods
(Piduru et al., 2012; Soydal et al., 2013; Annunziata Before examining the imaging sequelae of radio-
et al., 2014; Cho et al., 2015). This will be dis- embolization, it should be noted that several imag-
cussed in detail in the ensuing section. PET/CT ing findings on preprocedural imaging have been
also shows improved detection of certain hepatic found predictive of treatment response.
metastasis compared with CT alone and may aid Scintigraphic imaging of 99mTc macroaggre-
in 90Y treatment planning and patient selection gated albumin (MAA) administered during the
(Annunziata et al., 2014). The relative efficacy of 90Y mapping is performed primarily to evaluate the

PET/CT compared with MRI is less clear. A recent lung shunt fraction (LSF) and reduce the incidence
prospective study showed that diffusion-weighted of radiation pneumonitis. However, emerging evi-
MRI (DW-MRI) provided better evaluation of dence suggests that the LSF may yield important
early response of hepatic metastasis to 90Y than prognostic information not directly related to
PET/CT (Barabasch et al., 2015). However, these extrahepatic radiation dose deposition. A study
results should be taken in the context of the small of 62 patients with colorectal carcinoma (CRC)
patient series (n = 35) and its exclusion of HCC metastasis found that LSF was an independent pre-
from the study cohort (Barabasch et al., 2015). In dictor of survival after radioembolization. Patients
practice, the superiority of MRI over PET/CT is with LSF above the median value of 7.3% had
likely situational, depending on variable tumor significantly worse survival than those with LSF
histology and 18FDG avidity, as well as the ability below 7.3% (Deipolyi et al., 2014). This could reflect
to acquire high-quality MRI images. a phenomenon of increased systemic shunting of
Posttreatment imaging after radioembolization circulating tumor cells in patients with increased
conventionally begins 1 month after therapy, with LSF. Interestingly, hepatic tumor burden was not
284 The use of postprocedural imaging in the medical management of patients

associated with poor outcomes in this cohort 90Y therapy (Schlaak, 2013). Familiarity with the
(Deipolyi et al., 2014). constellation of possible imaging findings in the
Several imaging features on pretreatment CECT post-90Y patient is therefore important, so as to tai-
also correlate with survival. In patients with HCC, lor an appropriate follow-up regimen and triage to
increased central hypervascularity and well-defined adjuvant therapies when appropriate.
tumor margins are associated with improved sur-
vival (Salem et al., 2013a). In other words, infiltrative
and necrotic tumors infer relatively worse prognosis
in HCC. Similarly, centrally necrotic neuroendo- 14.2.3.1 0–3 months: rim
crine tumor (NET) metastases have also been asso- enhancement, necrosis, and
ciated with to poor response to 90Y therapy and poor pseudoprogression
prognosis (Neperud, 2013).
Changes in SUVmax as demonstrated on 18FDG- One of the earliest findings after radioembolization
PET/CT have been associated with early prediction is rim enhancement along the margins of the target
of treatment response after radioembolization. tumor (Singh and Anil, 2013). Rim enhancement
However, the volume of hypermetabolic tumor less than 5 mm in thickness is a common finding,
seen on preprocedural 18FDG-PET/CT has also occurring in approximately one-third of patients
been shown to be a solitary predictor of outcomes after 90Y (Keppke et al., 2007). This is in contradis-
in patients with unresectable hepatic metasta- tinction to normal imaging findings after TACE,
ses from melanoma (Piduru et al., 2012). In one where elimination of tumor vascularity is the
small series, patients with metabolic tumor burden desired treatment endpoint. For 90Y, it is important
(hypermetabolic tumor volume/total liver volume) to emphasize that the presence of thin rim enhance-
greater than 7% had markedly reduced prognosis. ment is not necessarily indicative of recurrent or
However, it is unclear whether this finding is recip- residual disease. Instead, this finding typically
rocated in other tumor histologies. reflects granulation tissue along the margins of the
treatment site. In fact, thin rim enhancement has
been reported in a high percentage of patients found
to have complete pathologic response to 90Y follow-
14.2.3 NORMAL RESPONSE TO ing transplant (Kulik et al., 2006). In once study of
THERAPY 46 patients with HCC, 80% of patients with thin
rim enhancement had response to therapy, whereas
Based on prior research, it can be reasonably 13% had stable disease (Keppke et al., 2007). Thin
assumed that a technically successful radioem- rim enhancement is usually transient, occurring
bolization therapy will result in some degree of between 1 and 2 months and resolving between 4
tumor response using standard dosimetry meth- and 5 months after therapy (Singh and Anil, 2013).
ods (Sangro et al., 2006; Sato et al., 2006; Salem Thick rim enhancement surrounding a lesion
et al., 2013b). However, the degree to which tumor after radioembolization is less specific. When
response occurs, the associated imaging findings, observed in the early posttreatment time frame,
and the time frame in which posttherapy changes this finding could reflect regional hyperemia
evolve are often variable. This is probably a result related to therapy (Figure 14.1). This is a particu-
of the unique mechanism of radioembolization, larly prevalent normal finding following radia-
which imparts both radiation and some degree of tion segmentectomy. However, persistence of
microvascular embolization in the treatment zone thick rim enhancement beyond 3 months or asso-
(Sangro et al., 2006; Sato et al., 2006; Salem et al., ciated nodularity is concerning for residual dis-
2013b). It should be emphasized that some stan- ease (Kulik et al., 2006). In another study, residual
dardized and widely accepted methods of report- nodular arterial phase enhancement seen on early
ing tumor response are optimized for reporting surveillance imaging (mean = 55 days) was asso-
response to cytotoxic agents rather than radiation. ciated with progressive disease in a high propor-
Consequently, World Health Organization (WHO) tion of patients (Keppke et al., 2007; Singh and
criteria and Response Evaluation Criteria in Solid Anil, 2013). In contrast, progressive low attenu-
Tumors (RECIST) may misrepresent the effect of ation within the treatment site usually indicated
14.2 Postprocedural imaging / 14.2.3 Normal response to therapy 285

mass effect and are usually distributed along


lesional margins or in a vascular distribution
(Miller et al., 2007). Like thin rim enhancement,
this finding may mimic residual tumor. However,
this finding is usually transient in nature, dis-
appearing around 3 months following therapy
(Miller et al., 2007; Singh and Anil, 2013) and is
postulated to reflect the same radiation-induced
inflammatory reaction commonly observed after
EBRT (Atassi et al., 2008a; Salem et al., 2013b;
Wang et al., 2013). However, as in TACE, a com-
ponent of microvascular occlusion may contrib-
ute to these altered enhancement characteristics
Figure 14.1 Thick rim enhancement is a nonspe- (Chung et al., 2010).
cific finding early after radioembolization, and Each of the aforementioned findings reflects
may be a normal finding. This finding was indica- normal hepatic changes of radioembolization
tive of regional hyperemia in this patient who
within the treatment size and may be seen on an
underwent radiation segmentectomy. Nodular
rim enhancement is a more worrisome finding. unpredictable basis. When tumoral/peritumoral
(Reproduced from Minocha, J. and Lewandowski, edema and peripheral enhancement are collec-
R., Semin. Intervent. Radiol., 28, 226–229, 2011. tively present in the early (<3 months) posttreat-
With permission.) ment period, it may be particularly difficult to
distinguish from disease progression. In this set-
necrosis, another common early imaging finding ting, lesional margins become indistinct and the
after 90Y. tumor may appear to increase in size, a phenome-
Approximately 95% of the radiation dose from non commonly referred to as “pseudoprogression”
90Y is delivered within four half-lives, or approxi- (Salem et al., 2013b; Singh and Anil, 2013; Dhingra
mately 11 days. Consequently, early changes of et al., 2014). However, it is important to note that
radiation-induced coagulative necrosis and asso- tumors with this appearance decrease in average
ciated peritumoral edema can be frequently attenuation, indicating necrosis and peritumoral
seen in the postprocedural period from 1 to 3 edema (Salem et al., 2013b; Dhingra et al., 2014).
months(Singh and Anil, 2013). Necrosis mani- This finding helps to distinguish from early pro-
fests as an area of low attenuation on CECT with gression of disease, usually denoted by increased
hypoenhancement on both CECT and MRI solid enhancing tumor (Salem et al., 2013b; Singh
(Keppke et al., 2007; Miller et al., 2007). It is and Anil, 2013; Salem et al., 2013b; Dhingra et al.,
important to recognize necrosis as an indicator of 2014). At times, progressive disease and pseudo-
early treatment response, as reduction in tumor progression can be hard to distinguish and imag-
size can take months to occur. In fact, median ing findings must be taken in context of serum
time to response has been demonstrated to be tumor markers, clinical status, and histologic risk
approximately 5–6 months using only size crite- factors. A follow-up exam at the 3- to 6-month
ria, compared with 1 month when using both size interval after 90Y can be helpful to confirm pseudo-
and necrosis as criteria (Keppke et al., 2007; Miller progression if reduction in tumor volume is noted.
et al., 2007; Singh and Anil, 2013; Salem et al.,
2013b). This is in keeping with the 3 to 6-month
delay that is generally expected to see reduction 14.2.3.2 Beyond 3 months: reduction
in tumor volume after external beam radiation in tumor size and changes in
therapy (EBRT) (Salem et al., 2013b). Patchy areas liver volume
of regional hypoenhancement not meeting crite-
ria for necrosis may also be transiently seen in the Reduction in tumor volume is an important end-
90Y treatment site. This finding has been noted on point in radioembolization (Singh and Anil, 2013).
portal venous phase CT in approximately 40% of In those patients who will experience a therapeu-
patients. These areas are without corresponding tic response, it is generally assumed that reduction
286 The use of postprocedural imaging in the medical management of patients

in tumor volume occurs in a delayed fashion after 14.2.3.3 Radiation lobectomy and
treatment, as seen in EBRT (Salem et al., 2013b; segmentectomy
Singh and Anil, 2013). While tumoral necrosis is
often visible 0–3 months after 90Y, reduction in Radiation lobectomy refers to the lobar infusion
tumor volume occurs more commonly from 4 to 6 of relatively high 90Y activity in patients with uni-
months (Miller et al., 2007) (Figure 14.2). It should lobar disease to provide the dual effect of treat-
also be noted that radiation changes within the ing the tumor and inducing contralateral lobe
unaffected liver lobe or segment that underwent hypertrophy (Gaba et al., 2009). This method
treatment also become visible at this time. has a demonstrated ability to induce ipsilateral
Although the unaffected liver tissue in the 90Y lobar atrophy and contralateral lobar hypertrophy
treatment zone generally receives significantly averaging 52% and 40%, respectively, comparable
reduced dose compared with neoplastic tissue, with portal vein embolization (Gaba et al., 2009).
NTE to normal liver may present variable imaging Further, patients treated with radiation lobectomy
sequelae on delayed (>3 months) follow-up imaging. have shown improved tumor response and sur-
Namely, changes in liver volume frequently occur, vival, with a comparable 5 years to surgery (36.6
including ipsilateral lobar atrophy and compensatory months) (Jakobs et al., 2008). The conceptual basis
contralateral hypertrophy (Jakobs et al., 2008). This of radiation lobectomy has also been applied in
phenomenon was examined in a cohort of patients a superselective fashion to a hepatic segment, in
who underwent glass radioembolization by variable patients with localized disease but contraindica-
infusion techniques (i.e., lobar, bilobar infusion). The tions to ablation or surgery. This technique has
authors noted an 11.8% decrease in total liver volume been referred to as “radiation segmentectomy”
following bilobar infusion. In patients undergoing and involves administration of high activity of 90Y
unilobartherapy, an ipsilateral volume loss of 8.9% to a localized area of the liver, allowing absorbed
and 21.2% increase in volume of the contralateral doses greater than 1000 times than those deliv-
lobe was noted (Jakobs et al., 2008). Hepatic atrophy ered in EBRT (Salem et al., 2013b). Subsequently,
from radioembolization is characterized histologi- the treated hepatic segment often experiences
cally by fibrosis, which may change enhancement significant atrophy and may disappear on follow-
characteristics on follow-up imaging. Although the up imaging. Additional information specific to
degree of hepatic atrophy can be significant, this radiation segmentectomy and lobectomy is avail-
effect is commonly asymptomatic (Singh and Anil, able in Chapter 6.
2013; Brown, 2014).

63.92 mm 42.97 mm
[R]
46.23 mm
56.57 mm R]

(a) (b)

Figure 14.2 (a) Coronal computed tomography (CT) showing large enhancing hepatocellular carci-
noma that was subsequently treated with radioembolization; (b) complete response to therapy follow-
ing radioembolization by modified Response Evaluation Criteria in Solid Tumors (mRECIST). Note that
in spite of complete tumor necrosis, minimal decrease in tumor size was noted. (Reproduced from
Singh, P. and Anil, G., Cancer Imag., 13, 2013 under the terms of the Creative Commons Attribution
4.0 International License; http://creativecommons.org/licenses/by/4.0/.)
14.2 Postprocedural imaging / 14.2.4 Follow-up imaging assessment criteria 287

14.2.3.4 Sequela of portal imaging sequelae of worsening portal hyperten-


hypertension after therapy sion can be seen using both resin and glass micro-
spheres and should be interpreted in context of
Although many patients may be asymptomatic, their clinical relevance and underlying functional
the delivery of significant radiation to the nor- hepatic reserve.
mal hepatic parenchyma can have clinical con- Low-volume perihepatic ascites is a common
sequence, as a result of fibrotic remodeling. Just early finding following radioembolization and
as lobar 90Y infusion may incite contralateral does not necessarily represent evidence of por-
lobar hypertrophy via induction of ipsilateral tal hypertension or worsening hepatic function.
fibrosis and atrophy, this method may exacerbate Instead, this is thought to result from irritation of
portal hypertension. This is particularly true in the Glisson capsule (Hilgard et al., 2010). A similar
patients with poor hepatic functional reserve. phenomenon may also occur in the adjacent lung
Consequences of worsened portal hyperten- base as a result of pleural irritation (Singh and
sion can often be seen in surveillance imaging Anil, 2013). In either case, findings are self-limited
(Lam et al., 2013a) (Figure 14.3). main portal and infrequently warrant intervention.
vein (MPV) diameter has been shown to increase
after both bilobar and unilobar infusion of glass
microspheres (Jakobs et al., 2008). In patients 14.2.3.5 Progression of disease
who received bilobar treatment, delayed follow-
up imaging showed 28% increase in splenic size, Disease progression manifests as new or enlarging
as well as increased diameter of the MPV, splenic hepatic tumor burden. It is often the result of new
vein, and superior mesenteric veins (Jakobs et al., tumor formation or growth of microscopic rests
2008). Not surprisingly, these imaging findings of tumor cells unlikely to be effected by radio-
correlate with worsening thrombocytopenia in embolization, or from suboptimal dose delivery
some patients as a result of hepatic sequestration to the tumor (Sangro et al., 2006). It is important
of platelets (Lam et al., 2013a). Increased number to note that median time to progression (TTP)
and size of porto-systemic collaterals vessels have occurs relatively late in patients without portal
also been noted following lobar infusion. These vein thrombus, appearing around 12–16 months
after therapy (Hilgard et al., 2010; Salem et al.,
2010). In all patients, TTP ranges from approxi-
mately 10–12 months (Hilgard et al., 2010; Salem
et al., 2010). The determination of disease progres-
sion is usually augmented clinical and serologic
information, as well as baseline tumor histologic
risk factors. Identifying early disease progression
is important, as triage to additional/alternative
liver-directed therapy or systemic agents may help
to prolong survival.

(a) (b)
14.2.4 FOLLOW-UP IMAGING
Figure 14.3 This image illustrates sequelae of ASSESSMENT CRITERIA IN
worsening portal hypertension following radio- PREDICTING TREATMENT
embolization. (a) Prior to radioembolization
and (b) 3 months postradioembolization. This
RESPONSE
57-year-old patient underwent bilobar therapy
The WHO criteria and RECIST offered the first
for colorectal metastases. Spleen volume
increased 84.8% after therapy and platelet vol- standardized methods of assessing the effect of
ume decreased 45.2%. (Reproduced from Lam, an oncologic therapy. Both of these guidelines
M.G.E.H. et al., Cardiovasc. Intervent. Radiol., 37, were initially optimized for reporting response
1009–1017, 2013a. With permission.) to systemic cytotoxic therapy, accounting only
288 The use of postprocedural imaging in the medical management of patients

for changes in tumor size (Lencioni and Llovet, enhancement in determining response. A sep-
2010). As locoregional therapies aimed at devas- arate assessment method described by Choi
cularizing liver tumors gained use, it became clear et al. for evaluating gastrointestinal stromal
that WHO and RECIST underestimate response tumors (GIST) incorporates changes in both
rates in HCC (Miller et al., 1981; Therasse et al., tumor size and mean tumor density (Choi et al.,
2000; Lencioni and Llovet, 2010) (Figure 14.4). 2007). The Choi criteria have since been applied
This is due in large part to the early appearance to the surveillance of HCC and liver metastases
of tumor necrosis in response to locoregional following radioembolization (Schlaak, 2013).
therapies such as radioembolization, a finding Each of these criteria can be used for both CT
that occurs months before reduction in tumor and MRI, and each is outlined in Figure 14.4 and
size (Figure 14.2). Assessment systems incorpo- Table 14.1. The primary question asked regard-
rating tumor enhancement characteristics were ing these various assessment methods is “which
therefore thought to more accurately represent one is most accurate in predicting response
TTP of liver tumors. Consequently, the WHO and time to progression?” This question is not
and RECIST criteria were modified in 2000 and trivial as palliation of liver malignancy hinges
2008, respectively, to account for tumor enhance- on month-to-month changes in disease status
ment characteristics. These new criteria, referred so as to tailor an effective treatment regimen to
to as the European Association for the Study of each patient. Fortunately, the use of each of the
the Liver (EASL) and modified RECIST (mRE- aforementioned criteria has been the subject of
CIST), and others such as the Choi criteria, have at least some comparative research. The basis
gained increasing utility in determining response for use of enhancement criteria (mRECIST and
to radioembolization. EASL) derives from experience with chemoem-
While a comprehensive discussion of the bolization. In a large cohort of patients undergo-
development and use of the various assessment ing TACE, mRECIST and EASL more accurately
criteria is outside of the scope of this chapter, predicted survival than WHO and RECIST cri-
it is important to understand their general dif- teria (Shim et al., 2012). Consequently, tumor
ferences. As previously discussed, the original enhancement gained favor over tumor size as
WHO and RECIST criteria account for only a primary determinant of treatment response
size. Whereas the WHO criteria include bidi- for chemoembolization and was then applied
rectional tumor size, RECIST accounts only for to radioembolization (Shim et al., 2012). The
longest tumor dimension. EASL and mRECIST Choi criteria, which incorporate mean tumor
criteria also account for arterial phase tumor enhancement and tumor size, have been shown

Approaches to response
measurement

Bidimensional Unidimensional

WHO EASL RECIST mRECIST


(Size criteria) (Enhancement) (Size criteria) (Enhancement)

Complete response: Total disappearance Complete response: Total disappearance


Partial response: 50% decrease Partial response: 30% decrease
Progressive disease: 25% increase or new lesions Progressive disease: 20% increase or new lesions
Stable disease: Neither PR or PD Stable disease: Neither PR or PD

Figure 14.4 Summary of assessment criteria for oncologic reporting.


14.2 Postprocedural imaging / 14.2.4 Follow-up imaging assessment criteria 289

Table 14.1 Comparison of WHO, RECIST, mRECIST, EASL, and Choi criteria

response WHO rECISt EaSL mrECISt Choi


Complete Disappearance Disappearance Disappearance Disappearance Disappearance
of all known of all target of all of any of all target
disease lesions enhancing intratumoral lesions
disease arterial
enhancement
in all target
lesions
Partial At least 50% At least a 30% At least 50% At least a 30% Decrease in
decrease in decrease in decrease in decrease in the tumor size
tumor size the sum of tumor sum of ≥10% or
from diameters of enhancement diameters of decrease in
baseline target from baseline viable tumor
lesions, (enhancement density
taking as in the arterial ≥15% on CT
reference the phase) target
baseline sum lesions, taking
of the as reference
diameters of the baseline
target lesions sum of the
diameters of
target lesions
Stable Any cases that Any cases that Any cases that Any cases that Any cases that
disease do not do not do not do not qualify do not
qualify for qualify for qualify for for either qualify for
either partial either partial either partial partial either partial
response or response or response or response or response or
progressive progressive progressive progressive progressive
disease disease disease disease disease
Progressive At least 25% An increase of At least 25% An increase of at Increase in
disease increase of at least 20% increased least 20% in the tumor size
one or more in the sum of enhancement sum of the ≥10% and
lesions, or the diameters of one or more diameters of does not
appearance of target lesions, or viable meet partial
of new lesions, appearance (enhancing) response
lesions taking as of new lesions target lesions, (PR) criteria
reference the taking as by tumor
smallest sum reference the density
of the smallest sum of
diameters of the diameters
target lesions of viable
recorded (enhancing)
since target lesions
treatment recorded since
started treatment
started
Notes: WHO, World Health Organization; RECIST, Response Evaluation Criteria in Solid Tumors; mRECIST, modified
RECIST; EASL, European Association for the Study of the Liver; CT, computed tomography.
290 The use of postprocedural imaging in the medical management of patients

to be superior to mRECIST in evaluating HCC widely used. A description of PERCIST is pro-


response to therapy in at least one single-center vided in Table 14.2.
review (Schlaak, 2013). However, it should be
noted that the Choi criteria were initially devel- 14.2.4.2 Investigational Follow-Up
oped for CT and include quantitative density
Methods
analysis specific to CT (Bonekamp et al., 2013)
that may be somewhat imprecise when used in While postradioembolization surveillance usually
MRI follow-up. involves evaluation of tumor size/enhancement
and FDG avidity when applicable, a number of
14.2.4.1 The Role of 18Fdg-Pet/Ct additional methods have been described on small
patient series. CT perfusion (CTp) involves serial
Although tumor enhancement is now primarily acquisition of CT images after administration
emphasized in evaluating effects of locoregional of iodinated contrast to characterize and quan-
therapy, follow-up assessment with 18FDG-PET/ tify enhancement. The general principle of its use
CT also plays a significant role after 90Y treat- relates to the importance of tumor vascularity as a
ment. PET/CT is particularly useful in surveil- prognostic factor after locoregional therapy (Shim
lance of metastatic disease to liver, which is et al., 2012), as previously discussed. However,
generally metabolically active (Singh and Anil, CTp allows for more accurate quantification of
2013; Vouche et al., 2015). For many metastatic enhancement than simple visual estimation.
tumors such as CRC, 18FDG-PET/CT has shown Preliminary studies using CTp have shown that
superiority to CT and MRI for lesional detection significant reduction in post-90Y perfusion trans-
and is similarly helpful in determining residual lates to treatment response of metastatic disease
viable tumor after 90Y (Sacks et al., 2011). Further, (Reiner et al., 2014). Interestingly however, these
18FDG-PET/CT may provide important prognos-
findings were not corroborated in patient with
tic information in the early posttreatment period HCC (Reiner et al., 2014).
after 90Y. Prior research has shown that quanti- In some situations, inflammatory changes
tative changes in SUVmax on 18FDG-PET/CT at from radioembolization and viable tumor may
6 weeks after therapy predict early response to be difficult to distinguish. One potential trouble-
radioembolization (Soydal et al., 2013). However, shooting method could be DWI-MRI. In a cohort
it should be emphasized that PET/CT should of 20 patients with HCC, quantitative analysis of
interpreted with caution prior to 2 months, so the ADC map on DWI-MRI predicted response
as to not confuse postprocedural inflammation within 42 days of radioembolization, 2 months
with residual/progressive tumor not seen on pre- prior to reduction in tumor size (Rhee et al.,
procedural imaging. The reporting scheme for 2008) (Figure 14.5). It remains unclear how these
18FDG-PET/CT is considerably less controversial
methods compare with mRECIST and EASL
than for CT and MRI, and the PET Response in larger patient cohorts and in variable tumor
Criteria in Solid Tumors (PERCIST) criteria are histology.

Table 14.2 PERCIST criteria for oncologic reporting


PErCISt criteria
Complete response Disappearance of all metabolically active lesions
Partial response 30% and 0.8-unit decline in (SUL SUV normalized to lean body
mass and corrected to background) peak between the most
intense lesion before treatment and the most intense lesion
after treatment, although not necessarily the same lesion
Progressive disease 30% and 0.8-unit increase in SUL peak or new lesions or 75%
increase in total lesion glycolysis
Stable disease Neither partial response nor progressive disease.
Note: PERCIST, Positron Emission Tomography Response Criteria in Solid Tumors.
14.3 Imaging of complications / 14.3.1 Hepatic toxicity 291

(a) (b) (c)

(d) (e) (f)

Figure 14.5 Images illustrating the utility of contrast-enhanced and diffusion-weighted imaging on
magnetic resonance imaging (DWI-MRI) in the evaluation of treatment response: (a) a metastatic
lesion was noted in the right hepatic lobe prior to therapy by CT. (b and c): Lesion has increased in
size 1 month after radioembolization. The treatment site manifests as (d) hypodensity on CT and
(e) peripherial nodular enhancement by contrast-enhanced MRI. (f) The absence of signal on DWI-
MRI confirms the absence of residual disease after therapy. (Reproduced from Singh, P. and Anil,
G., Cancer Imag., 13, 645–657, 2013 under the terms of the Creative Commons Attribution 4.0
International License; http://creativecommons.org/licenses/by/4.0/.)

radiation-induced liver disease (RILD) seen in


14.3 IMaGING OF EBRT for liver tumors and in whole-body radia-
COMPLICatIONS tion performed in preparation for allogenic bone
marrow transplant (Salem et al., 2013b). As one
Potential adverse events to the liver and extra- would expect, REILD presents several weeks after
hepatic soft tissues from 90Y radioembolization radiation dose delivery, commonly in the 4- to
are numerous and are often present with distinct 8-week posttherapy interval (Kuo et al., 2014).
clinical signs/symptoms. This section emphasizes Histologically, it is similar to RILD, characterized
imaging findings, pathogenesis, and epidemiol- by sinusoidal congestion, cholestasis, and areas
ogy of the more common complications from 90Y. of perivenular necrosis reflecting venooclusive
Complications that are not directly caused by 90Y disease (Sangro et al., 2008). REILD represents
microspheres such as vascular injury, exposure to a spectrum of disease and its clinical impact is
ionizing radiation, and iodinated contrast expo- generally determined by pretreatment hepatic
sure are also omitted from this discussion. functional reserve, the volume of tissue affected,
systemic chemotherapy, and presence of clini-
14.3.1 HEPATIC TOXICITY cal intervention. As expected, this phenomenon
occurs more frequently in patients treated in a
Radioembolization-induced liver disease (REILD) bilobar fashion, those who have undergone prior
is a form of hepatic subacute liver injury from EBRT, and in sequential radioembolization thera-
radiation exposure. It is conceptually similar to pies (Lam et al., 2013b). In the most severe cases,
292 The use of postprocedural imaging in the medical management of patients

and cholecystitis are much more common than


those related to extrahepatic NTE (Singh and
Anil, 2013). In one large series, combined bili-
ary complications occurred in approximately
10% of patients, requiring intervention in ~2%
(Atassi et al., 2008b). These rates are generally
higher in patients with prior biliary intervention
or biliary-enteric anastomosis (Riaz, 2014). This
relatively high rate of biliary toxicity is explained
by the vascular supply of the biliary tree. Unlike
normal hepatic parenchyma, the biliary tree lacks
a dual blood supply (Northover and Terblanche,
1979). Instead, the supraduodenal common bile
duct and hepatic duct are supplied by either the
gastroduodenal artery (68%) or right hepatic
Figure 14.6 Fulminant hepatic failure is largely artery (32%) (Northover and Terblanche, 1979).
a clinical diagnosis with nonspecific imaging The intrahepatic biliary tree is supplied by its
findings. This patient underwent lobar radio- accompanying lobar artery and its arterial vas-
embolization after left lobectomy for colorectal cular plexus (Northover and Terblanche, 1979).
carcinoma (CRC) metastasis. Eight weeks after This paucity of collateral vessels makes the biliary
therapy, the patient was found to have fulminant tree particularly susceptible to ischemic injury.
hepatic failure, manifested by patchy hypoen- Interestingly, peribiliary vascular hypertrophy
hancement. (Reproduced from Hamoui, N. and
found in cirrhotic livers is thought to prevent
Ryu, R., Semin. Intervent. Radiol., 28, 246–251,
2011. With permission.) peribiliary microsphere deposition and reduce
rates of biliary injury (Yu et al., 2002).
REILD can lead to fulminant hepatic failure and
death (Hamoui and Ryu, 2011). The overall inci- 14.3.2.1 Biloma
dence of REILD from radioembolization ranges
from approximately 4% to 9%, although it can The potential for biloma formation has been well
reach 50% in patients receiving aggressive che- documented in patients undergoing chemoemboli-
motherapy regimens (Sangro et al., 2008). REILD zation for hepatic malignancy, occurring in approx-
is primarily diagnosed by its clinical features; it imately 4% of all cases (Minocha and Lewandowski,
has been accompanied by ill-defined heterogenous 2011). Biloma are most commonly caused by small
hypoenhancement on short-term follow-up MRI vessel ischemia resulting in bile duct disruption,
and CT (Hamoui and Ryu, 2011) (Figure 14.6). although radiation effects may also contribute.
Hepatic toxicity from90Y is discussed in more Comparative rates of biloma in TACE and 90Y are
detail in Chapter 8, including a comprehensive unclear as are incidence rates among the glass and
discussion of the hepatic dose toxicity models. resin products. However, a large independent obser-
vational study reported rates approximating 1% for
radioembolization. On imaging, biloma typically
14.3.2 BILIARY EFFECTS manifests as a well-defined intrahepatic collec-
tion approximating the biliary tree (Figure 14.7). A
While REILD is a serious concern that greatly direct communication with the bile ducts may not
effects 90Y treatment planning and much tech- be seen by CT or MRI, but is usually demonstrated
nical emphasis is placed on avoiding complica- by cholangiogram (Figure 14.8). The presence of rim
tions from NTE, biliary toxicity provides some enhancement is common and should be considered
of the most common adverse effects of radioem- suspicious for secondary infection only in the clini-
bolization (Atassi et al., 2008b; Singh and Anil, cal context of fever, right upper quadrant pain, and
2013). With current techniques, it is now gener- leukocytosis (Singh and Anil, 2013). In this setting,
ally assumed that biliary complications including biloma may difficult to differentiate from intrahe-
radiation cholangitis, biloma, biliary stricture, patic abscess.
14.3 Imaging of complications / 14.3.2 Biliary effects 293

(a) (b)

Figure 14.7 (a) CT scan prior to radioembolization shows multiple hepatic metastatic lesions. (b) Two
months after therapy, the right hepatic lobe shows significant volume loss and capsular retrac-
tion with several hypodense lesions typical of intrahepatic biloma. (Reproduced from Singh, P. and
Anil, G., Cancer Imag., 13, 645–657, 2013 under the terms of the Creative Commons Attribution 4.0
International License; http://creativecommons.org/licenses/by/4.0/.)

(a) (b)

Figure 14.8 A 59-year-old male patient underwent radioembolization for melanoma metastasis
to liver. (a) Axial T1-weighted, fat-saturated MRI image 25 weeks after therapy shows nonenhanc-
ing lesions with thick rim enhancement in the right hepatic lobe and multiple left lobe metastasis.
(b) Administration contrast through a percutaneous drainage catheter demonstrates communication
with the biliary tree, confirming intrahepatic biloma. (Reproduced from Atassi et al., Radiographics,
28, 81–99, 2008a. With permission.)

14.3.2.2 Cholangitis and transient distinguished from worsening painless jaundice.


hyperbilirubinemia Grade III or higher bilirubin toxicity (total bili-
rubin >3.0 mg/dL) has been reported in as many
Radiation cholangitis is a clinical syndrome mani- as one-third of patients after therapy (Smits et al.,
fested by jaundice, fever, and right upper quadrant 2013). These findings are thought to more likely
pain. It is rare after radioembolization, with unclear represent effects of REILD than biliary toxicity
incidence and no clearly defined characteristic and may improve with conservative management
imaging findings (Riaz, 2014). While radiation (Salem et al., 2013b). It should be emphasized that
cholangitis may be sterile, it is commonly treated isolated mild hyperbilirubinemia is commonly
with antibiotics (Riaz, 2014). This should be asymptomatic and not associated with significant
294 The use of postprocedural imaging in the medical management of patients

changes in synthetic liver function or impend- leukocytosis. In clinical practice, rates of radia-
ing hepatic decompensation (Salem et al., 2013b; tion cholecystitis are highly variable, depending
Smits et al., 2013). on technical considerations such as prophylactic
embolization of the cystic artery and use of anti-
14.3.2.3 Biliary stricture reflux devices (Pasciak et al., 2015). Many inter-
ventional radiologists do not routinely embolize
Biliary stricture with upstream dilation often the cystic artery to prevent NTE to the gallblad-
reflects chronic sequela of bile duct injury on the der when it is downstream to the infusion site.
same spectrum biloma, potentially due to radiation This is due in large part to the possible risk of
and/or ischemia, which most commonly effects ischemic cholecystits from prophylactic cystic
intrahepatic biliary radicals and is seen to a lesser artery embolization, thought by many to out-
degree in cirrhotic livers. Many of these patients weigh the risk of gallbladder NTE (Hickey and
will be asymptomatic, only requiring intervention Lewandowski, 2011). In situations where the cys-
if clinical symptoms present as biliary obstruction tic artery is likely to sump a significant quantity
or secondary infection (Atassi et al., 2008a, 2008b; of 90Y activity from the tumor, or when signifi-
Singh and Anil, 2013). cant deposition of MAA is seen on the pretreat-
ment simulation, prophylactic embolization can
14.3.2.4 Radiation cholecystits be considered. Caution should be taken in embo-
lizing a dominant cystic artery that comprises all
Radiation cholecystitis is a clinical syndrome or nearly all blood flow to the gallbladder, as this
of cholecystits in patients recently undergoing is associated with increased risk of perforation
yttrium-90 radioembolization. Imaging findings (Hickey and Lewandowski, 2011). Fortunately,
of cholecystitis have been reported in approxi- radiation cholecystitis is usually self-limited and
mately 2% of patients, manifested by gallblad- managed with conservative therapy. Rarely, gall-
der wall thickening and hyperenhancement bladder perforation and need for surgical cho-
(Sag et al., 2014) (Figure 14.9). These imaging lecystectomy have been reported, but both are
findings accompany the clinical finding of right thought to occur in less than 1% of cases (Atassi
upper quadrant pain, with or without fever and et al., 2008b).

(a) (b)

Figure 14.9 (a) Contrast-enhanced CT scan performed 3 days after radioembolization demonstrates
gallbladder wall thickening and pericholecystic fluid suggestive of cholecystitis. The patient underwent
laparoscopic cholecystectomy 7 weeks later. (b) Microscopic analysis of the surgically removed gallblad-
der showed fibrosis, chronic inflammation, and the presence of glass microspheres (arrow). (Reproduced
from Hickey, R. and Lewandowski, R., Semin. Intervent. Radiol., 28, 230–233, 2011. With permission.)
14.3 Imaging of complications / 14.3.4 Gastrointestinal nontarget embolization 295

14.3.3 HEPATIC ABSCESS containing residual radioactivity. Ulceration is


commonly caused by reflux of microspheres into
Any rim-enhancing intrahepatic fluid collection the gastroduodenal (GDA) or right gastric arteries
should be suspicious for abscess in a patient after (RGA), where they may deposit and cause hemor-
radioembolization. The presence of internal gas rhage, inflammation, and/or ulceration (Veloso
raises diagnostic probability; however, it should be et al., 2013; Baumann et al., 2015). Occasionally,
emphasized that loculated foci of gas may be pres- peripheral deposition of 90Y within the liver can
ent in the liver normally after treatment. This is a result in clinically significant dose deposition to
result of low-volume, intra-arterial gas injection adjacent visceral structures (Singh and Anil, 2013).
during 90Y infusion, when it can become trapped Radiation-induced injury to mucosal stem cells
between microspheres for several days before is thought to be the primary causative etiology,
resorbing (Singh and Anil, 2013). Therefore, imag- with ischemia effects playing a lesser role. Because
ing findings suggestive of abscess should be taken mucosal stem cells are permanently depopulated
in context of clinical presentation. Percutaneous by radiation effects, radiation-induced GI tract
aspiration for culture may prove definitive in cases ulcers rarely resolve spontaneously. In order to
of unclear imaging and clinical findings (Singh mitigate ulceration risks, prophylactic emboliza-
and Anil, 2013). tion of the GDA and RGA, antacid medicine, and/
Hepatic abscess may occur after radioem- or use of antireflux devices may employed (Pasciak
bolization as a result of bacterial colonization et al., 2015). However, the practice of routine
of necrotic tumor, necrotic nontarget hepatic GDA embolization has been called into question
parenchyma, or biloma. Because ascending in light of recent research and is performed based
infection through the biliary tree is the most on institution-specific preference (Haydar et al.,
common route of infection, rates of hepatic 2010). The exact toxicity thresholds to the small
abscess are increased in patients with prior bowel and stomach from 90Y are unclear, although
sphincerotomy or biliary-enteric anastomosis. concurrent systemic chemotherapy likely sensi-
In patients with an intact sphincter of Oddi, tizes mucosal cells to radiation, increasing risk of
abscess occurs in approximately 1%–2% of cases ulceration (Brown et al., 2012).
(Brown et al., 2012). However, as many as 15% By imaging, radiation-induced GI tract ulcers
of patients with prior biliary-enteric anastomo- are indistinguishable from other forms of ulcer-
sis, sphincerotomy, or indwelling biliary stent ation, and may manifest as focal mural thicken-
experience an abscess, even when periprocedural ing, regional inflammatory changes, and/or signs
antibiotic were administered (Brown et al., 2012). of perforation. Endoscopy is the gold standard for
An aggressive antibiotic course combined with a diagnosis and should be considered in patients
bowel prep regimen has been shown to reduce, with any level of suspicion for NTE to bowel (Singh
and potentially eliminate, this risk (Cholapranee and Anil, 2013).
et al., 2014). Finally, radioembolization should be Acute pancreatitis is another rare sequela of
deferred in patients with suspicion for bactere- NTE observed in fewer than 1% of patients (Brown
mia, as subsequent abscess formation has been et al., 2012; Riaz, 2014). Pancreatitis manifests
reported (Mascarenhas et al., 2011). similarly when caused by radioembolization as
it does when caused by other etiologies, with ele-
14.3.4 GASTROINTESTINAL vated serum amylase/lipase and intense epigastric
NONTARGET pain. Imaging findings of acute pancreatitis related
EMBOLIZATION to 90Y are usually localized to the pancreatic head,
rather than diffuse in nature (Singh and Anil,
Gastrointestinal (GI) tract ulceration from NTE 2013). Postprocedural 90Y single-photon emission
is one of the most feared complications of radio- computed tomography (SPECT/CT) or PET/CT
embolization due to its morbidity and the com- imaging may be helpful to confirm microsphere
plex nature of surgical intervention in patients deposition in the pancreas and exclude other
potential causative etiologies.
296 The use of postprocedural imaging in the medical management of patients

14.3.5 RADIATION PNEUMONITIS cases of radiation pneumonitis were seen (Salem


AND OTHER SITES et al., 2008). The rarity of RP makes it subopti-
OF NONTARGET mally studied with respect to risk factors and
EMBOLIZATION optimal prevention strategies.
Nontarget embolization has been described
Radiation pneumonitis (RP) is the development in other unusual locations such as colon and
of pulmonary fibrosis and restrictive lung dis- kidney (Kao, 2014). These represent rare sites of
ease from exposure to sufficient radiation dose. NTE, with poorly understood clinical outcomes
It manifests in a similar fashion to organizing as a result of their rarity. Of particular interest,
pneumonia or chronic eosinophilic pneumonia NTE to the abdominal wall from a falciform
on CT, with patchy areas of ground-glass opac- artery has been reported. The falciform artery
ity, mild volume loss, and traction bronchiectasis is a small terminal arterial branch off the left
(Singh and Anil, 2013) (Figure 14.10). Making hepatic or proper hepatic arteries that extends to
the diagnosis RP is particularly important in the umbilicus and communicates with the epi-
patients being considered for repeat radioembo- gastric arteries (Bhalani and Lewandowski, 2011)
lization as the increased cumulative dose could (Figure 14.11). Failure to recognize and embolize
worsen pulmonary function (Wright et al., 2012). potential NTE to abdominal wall from a falci-
Avoiding RP is a significant emphasis in pre- form artery can result in periumbilical abdomi-
therapy treatment planning, and is the basis for nal pain, cutaneous burning sensation, or skin
MAA administration and calculation of shunt necrosis (Bhalani and Lewandowski, 2011; Smith
fraction. Dosimetric and radiobiologic consid- et al., 2015). However, it has recently been shown
erations regarding RP are discussed in further that topically applied ice to the abdominal wall
detail in Chapter 4. Although the avoidance of in the periprocedural period can induce suffi-
RP is greatly emphasized in 90Y treatment plan- cient superficial vasoconstriction to eliminate
ning, its incidence is exceedingly rare. It could this risk.
be argued that rigorous treatment planning helps
to avoid RP; however, it is rarely reported even
in cases in which the lungs receive the assumed 14.4 POSttrEatMENt 90Y
toxicity threshold of 50 Gy. In one cohort of 58 IMaGING
patients receiving a lung dose above 50 Gy no

14.4.1 EVALUATING NONTARGET


EMBOLIZATION WITH
POSTTREATMENT 90Y
IMAGING
Recall that there are no reliable angiographic
methods to ensure target delivery of micro-
spheres and exclude nontarget embolization.
Both glass and resin microspheres are radiolucent
and thus not visualized on fluoroscopy. Although
dilute iodinated contrast is commonly used to
flush the delivery catheter and confirm antero-
grade flow during infusion of resin microspheres,
this method provides limited evaluation for NTE.
Figure 14.10 Axial thoracic CT showed diffuse It is also known that 90Y microspheres have the
ground glass opacity with traction bronchiectasis
potential to distribute in an unpredictable fash-
in a patient following radioembolization. Given
this appearance and progressive nature, this was ion compared with that of the preprocedural
determined to be a result of radiation pneumoni- Tc99m-MAA injection, owing to differences in
tis. (Reproduced from Wright, C.L. et al., J. Vasc. particle size, catheter position, injection rate,
Interv. Radiol., 23, 2012. With permission.) presence of stasis, and changes in tumor vascular
14.4 Posttreatment 90Y imaging / 14.4.2 Prognostication with 90Y PET/CT 297

(a) (b)

Figure 14.11 (a) Pretreatment angiography prior to radioembolization in a 57-year-old patient with
CRC metastases to liver. Selective angiogram of the segment 4 branch demonstrated the presence of
a falciform artery (arrows). However, its diminutive size prevented coil embolization. After treatment,
the patient experienced localized supraumbilical pain thought to be related to nontarget emboliza-
tion of yttrium-90. (b) Left hepatic arteriogram prior to therapy in the same patient. (Reproduced
from Bhalani, S. and Lewandowski, R., Semin. Intervent. Radiol., 28, 234–239, 2011. With permission.)

dynamics (Lam, 2013c; Wondergem, 2013). exact tumor toxicity threshold remains incom-
Consequently, methods of postprocedural imag- pletely understood. Many factors contribute to
ing that allow for visualization of 90Y distribution this uncertainty, including variable methods
play an indispensible role in confirming technical of dose calculation, tumor heterogeneity, and
success and ensuring patient safety. intrinsic differences among the two 90Y micro-
Identification of GI and other NTE on posttreat- sphere products (Kao et al., 2013). Much of the
ment imaging is covered in detail in Chapter 13, in prior dose–response data for HCC and metas-
particular, the use of bremsstrahlung SPECT and tases derives from predictive dosimetry from
90Y PET/CT to gauge technical success and detect Tc99M-MAA SPECT/CT (Eaton et al., 2014;
NTE, which is not straightforward due to signifi- Kokabi et al., 2014). These studies have shown
cant differences between 90Y and conventional tumor-response threshold ranging from 120 Gy
diagnostic radionuclides. in HCC to 50 Gy in certain metastases (Dezarn
et al., 2011). While predictive dosimetry is fea-
sible and commonly used, it is hindered by the
14.4.2 PROGNOSTICATION WITH differences between 90Y microspheres and MAA
90Y PET/CT discussed previously. Therefore, dosimetry based
on Tc99M-MAA simulation provides only an
Survival and response to radioembolization is estimation of dose distribution that is likely to
multifactorial, depending on nontreatment- have been delivered to a tumor (Kao et al., 2013).
related factors such as tumor burden, histologic Other dosimetry methods predicated on brems-
subtype, systemic radiosensitizers, and others strahlung SPECT are further limited in their
(Gunduz et al., 2014). When solely focusing on accuracy.
tumor response, the amount of radiation deliv- The most exciting aspect of 90Y PET/CT is
ered to the tumor (tumor absorbed dose) plays the ability to obtain accurate tumor dosim-
a primary role. However, despite nearly two etry, as previously discussed in Chapters 11
decades of experience with radioembolization, and 13. 90Y PET/CT has the capability to yield
298 The use of postprocedural imaging in the medical management of patients

potentially important dose–response data and outcomes immediately following 90Y radioem-
prognostic information. According to phantom bolization. Figure 14.12 describes an example
studies, quantification of 90Y activity could be of the utilization of PET/CT in the prognostica-
performed with accuracy ranging from 0.4% to tion of a poor clinical response to therapy. Such
10% depending on scan parameters, significantly outcomes are not uncommon in radioemboliza-
better than SPECT/CT in comparative studies tion and can stem from relative hypovascularity
(Lhommel et al., 2009; Elschot et al., 2013; Gates of tumor, poorly optimized treatment planning,
et al., 2013). A recent international, multicenter and numerous technical and patient-specific
trial confirmed the consistent accuracy of 90Y challenges occurring during infusion. However,
dosimetry using modern PET/CT imaging with early prognostication of these outcomes can
time-of-flight (ToF) on a number of scanners allow for consideration of alternative and adju-
with variable scan parameters (Willowson et al., vant therapies several months earlier than was
2015). As quantitative dosimetry with PET/CT previously possible. The potential benefit of
has increased in scope, a more in-depth under- using quantitative posttreatment 90Y imaging in
standing of tumor response will likely come to the clinical management of disease, particularly
light. Such information, combined with 90Y PET/ in the context of terminal patients, cannot be
CT dosimetry, will allow for prognostication of overstated.

(a) (b)

(c) (d)

Figure 14.12 A patient with multifocal CRC liver metastases is treated with 1300 MBq of 90Y resin
microspheres. (a) Lesion appears hypodense on pretreatment contrast-enhanced CT (CECT), with an
SUVmax of 7.8 2 weeks prior to treatment as shown in (b). (c) Posttreatment quantitative 90Y positron
emission tomography/CT (PET/CT) reveals an average lesion dose of 64 Gy. (d) Follow-up PET/CT
at 8 weeks following treatment shows an SUVmax of 8.0 with no apparent therapeutic effect owing to
the low absorbed dose delivered to the tumor.
References 299

for a second infusion on the same day. The net result


14.5 trEatMENt MODIFICatION was a robust partial treatment response on follow-up
WItH 90Y PEt/Ct imaging. While the logistics of routinely using this
method make it prohibitive for routine use, it dem-
14.5.1 INTERPROCEDURAL onstrated that 90Y PET/CT may also have value in
TREATMENT MODIFICATION allowing salvage of a procedural with similar techni-
cal complications.
Yttrium-90 PET/CT dosimetry data have also been
used in clinical practice to tailor treatments in vari-
ous case reports. The first example of this was pub- 14.6 CONCLUSIONS
lished by Chang et al. (2013). In this case, a patient
with cholangiocarcinoma was treated with right
lobar infusion of resin microspheres for unresectable Imaging plays an important role in the plan-
recurrent disease after Whipple and radiofrequency ning, delivery, follow-up, and clinical manage-
ablation. Treatment planning was performed using ment of patients undergoing radioembolization.
the standard body surface area approach. The patient Familiarity with common complications and
experienced minimal (<25%) response to radioem- associated imaging findings may improve clinical
bolization on follow-up FDG-PET/CT. Review of outcomes. Postprocedural imaging with brems-
the 90Y PET/CT performed immediately after radio- strahlung SPECT and PET/CT can gauge early
embolization yielded a tumor-absorbed dose of treatment response and identify sites of nontarget
70 Gy and tumor to normal uptake ratio (T/N) of embolization. PET/CT is likely to play an impor-
2.5:1. For a subsequent left lobar infusion, this low tant role in refining the safety and efficacy profile
calculated T/N was used to justify increased activ- of radioembolization.
ity infusion that would approach toxicity thresholds
reported for HCC (Strigari et al., 2010). The left lobe REFERENCES
tumor received 110 Gy on postprocedural 90Y PET/
CT and achieved a complete response to therapy. Annunziata, S., Treglia, G., Caldarella, C.,
While this dosimetry method is somewhat time con- Galiandro, F. (2014). The role of 18F-FDG-PET
suming, it is similar to partition model dosimetry and PET/CT in patients with colorectal liver
using Tc99m-MAA and represents a step toward the metastases undergoing selective internal
refinement of dosimetry calculation and treatment radiation therapy with yttrium-90: a first
personalization. evidence-based review. Sci World J 2014 :1–8,
Article ID 879469. doi: 10.1155/2014/879469.
14.5.2 INTERPROCEDURAL eCollection 2014.
Atassi, B. et al. (2008a). Multimodality imag-
TREATMENT MODIFICATION
ing following 90Y radioembolization: A
Another technique of treatment modification comprehensive review and pictorial essay.
using 90Y PET/CT was performed in a similar fash- Radiographics 28:81–99.
ion, except both infusions targeted a single lesion Atassi, B. et al. (2008b). Biliary sequelae fol-
and the two infusions were performed on the same lowing radioembolization with yttrium-90
day (Bourgeois et al., 2015). In this case, a patient microspheres. J Vasc Interv Radiol
with HCC was treated with resin 90Y microspheres 19:691–697.
via right lobar infusion using the body surface area Attenberger, U.I. et al. (2015). Fifty years of
(BSA) dosimetry method. However, due to intrapro- technological innovation: Potential and limita-
cedural technical complications, only a fraction of tions of current technologies in abdominal
the 90Y was administered. The patient was promptly magnetic resonance imaging and computed
transferred to have a 90Y PET/CT, which showed 52 tomography. Invest Radiol 50:584–593.
Gy average tumor dose. A simple arithmetic con- Barabasch, A. et al. (2015). Diagnostic accuracy of
version was used to calculate the needed activity to diffusion-weighted magnetic resonance imag-
reach 120 Gy tumor dose and the patient returned ing versus positron emission tomography/
300 The use of postprocedural imaging in the medical management of patients

computed tomography for early response Cho, E. et al. (2015). 18F-FDG PET CT as a prog-
assessment of liver metastases to y90- nostic factor in hepatocellular carcinoma. Turk
radioembolization. Invest Radiol 50:409–415. J Gastroenterol 26:344–350.
Bargellini, I. (2014). How does selective inter- Choi, H. et al. (2007). Correlation of computed
nal radiation therapy compare with and/or tomography and positron emission tomog-
complement other liver-directed therapies. raphy in patients with metastatic gastroin-
Future Oncol 10:105–109. testinal stromal tumor treated at a single
Baumann, J., Lin, M., Patel, C. (2015). Image institution with imatinib mesylate: Proposal of
of the month. Clin Gastroenterol Hepatol new computed tomography response criteria.
13:A23–A24. J Clin Oncol 25:1753–1759.
Bester, L. et al. (2014). Transarterial chemoem- Cholapranee, A. et al. (2014). Risk of liver
bolisation and radioembolisation for the abscess formation in patients with prior bili-
treatment of primary liver cancer and second- ary intervention following yttrium-90 radio-
ary liver cancer: A review of the literature. J embolization. Cardiovasc Intervent Radiol
Med Imaging Radiat Oncol 58:341–352. 38:397–400.
Bhalani, S., Lewandowski, R. (2011). Chung, J. et al. (2010). Haemodynamic events
Radioembolization complicated by nontarget and localised parenchymal changes following
embolization to the falciform artery. Semin transcatheter arterial chemoembolisation for
Intervent Radiol 28:234–239. hepatic malignancy: Interpretation of imaging
Boas, F.E. et al. (2015). Optimal imaging surveil- findings. Br J Radiol 83:71–81.
lance schedules after liver-directed therapy Deipolyi, A.R. et al. (2014). High lung shunt
for hepatocellular carcinoma. J Vasc Interv fraction in colorectal liver tumors is associ-
Radiol 26:69–73. ated with distant metastasis and decreased
Bonekamp, S. et al. (2013). Unresectable survival. J Vasc Interv Radiol 25:1604–1608.
hepatocellular carcinoma: MR imaging after Dezarn, W.A. et al. (2011). Recommendations
intraarterial therapy. Part I. Identification and of the American Association of Physicists in
validation of volumetric functional response Medicine on dosimetry, imaging, and quality
criteria. Radiology 268:420–430. assurance procedures for 90Y microsphere
Bourgeois, A.C. et al. (2015). Intraprocedural brachytherapy in the treatment of hepatic
yttrium-90 positron emission tomography/ malignancies. Med Phys 38:4824.
CT for treatment optimization of yttrium-90 Dhingra, S. et al. (2014). Histological changes
radioembolization. J Vasc Interven Radiol in nontumoral liver secondary to radioem-
25:271–275. bolization of hepatocellular carcinoma with
Braat, A.J.A.T. et al. (2014). Hepatic radioem- yttrium 90-impregnated microspheres:
bolization as a bridge to liver surgery. Front Report of two cases. Semin Liver Dis
Oncol 4:199. 34:465–468.
Brown, K.T. (2014). Superselective yttrium-90 Eaton, B.R. et al. (2014). Quantitative dosimetry
radioembolization for hepatocellular for yttrium-90 radionuclide therapy: Tumor
carcinoma in high-risk cases: Another dose predicts fluorodeoxyglucose positron
tool in the toolbox. J Vasc Interv Radiol emission tomography response in hepatic
25:1073–1074. metastatic melanoma. J Vasc Interv Radiol
Brown, D.B. et al. (2012). Quality improvement 25:288–295.
guidelines for transhepatic arterial chemo- Eldredge-Hindy, H. et al. (2014). Yttrium-90
embolization, embolization, and chemothera- microsphere brachytherapy for liver metas-
peutic infusion for hepatic malignancy. J Vasc tases from uveal melanoma. Am J Clin Oncol
Interv Radiol 23:287–294. 39:189–195.
Chang, T.T., Bourgeois, A.C., Balius, A.M., Elschot, M. et al. (2013). Quantitative compari-
Pasciak, A.S. (2013). Treatment modification son of PET and bremsstrahlung SPECT for
of yttrium-90 radioembolization based on imaging the in vivo yttrium-90 microsphere
quantitative positron emission tomography/ distribution after liver radioembolization, Villa
CT imaging. J Vasc Interv Radiol 24:333–337. E, ed. PLoS One 8:e55742.
References 301

Gaba, R.C. et al. (2009). Radiation lobectomy: brachytherapy: A Consensus Panel Report
Preliminary findings of hepatic volumetric from the Radioembolization Brachytherapy
response to lobar yttrium-90 radioemboliza- Oncology Consortium. Int J Radiat Oncol
tion. Ann Surg Oncol 16:1587–1596. Biol Phys 68:13–23.
Gates, V.L., Salem, R., Lewandowski, R.J. (2013). Keppke, A.L. et al. (2007). Imaging of hepa-
Positron emission tomography/CT after tocellular carcinoma after treatment
yttrium-90 radioembolization: Current and with yttrium-90 microspheres. AJR Am J
future applications. J Vasc Interv Radiol Roentgenol 188:768–775.
24:1153–1155. Khan, A.S. (2014). Current surgical treatment
Gunduz, S. et al. (2014). Yttrium-90 radio- strategies for hepatocellular carcinoma in
embolization in patients with unresect- North America. WJG 20:15007.
able liver metastases: Determining the Kim, H.J. et al. (2015). Incremental value of
factors that lead to treatment efficacy. liver MR imaging in patients with poten-
Hepatogastroenterology 61:1529–1534. tially curable colorectal hepatic metastasis
Hamoui, N., Ryu, R. (2011). Hepatic radioembo- detected at CT: A prospective comparison
lization complicated by fulminant hepatic of diffusion-weighted imaging, gadoxetic
failure. Semin Intervent Radiol 28:246–251. acid–enhanced MR imaging, and a combi-
Haydar, A., Wasan, H., Wilson, C., Tait, P. (2010). nation of both MR techniques. Radiology
90Y radioembolization: Embolization of 274:712–722.
the gastroduodenal artery is not always Kokabi, N. et al. (2014). A simple method for
appropriate. Cardiovasc Intervent Radiol estimating dose delivered to hepatocellular-
33:1069–1071. carcinoma after yttrium-90 glass-based radio-
Hickey, R., Lewandowski, R. (2011). Hepatic embolization therapy: Preliminary results of
radioembolization complicated by radia- a proof of concept study. J Vasc Interv Radiol
tion cholecystitis. Semin Intervent Radiol 25:277–287.
28:230–233. Kulik, L.M. et al. (2006). Yttrium-90 microspheres
Higgins, M., Soulen, M. (2013). Combining (TheraSphere) treatment of unresectable
locoregional therapies in the treatment of hepatocellular carcinoma: Downstaging to
hepatocellular carcinoma. Semin Intervent resection, RFA and bridge to transplantation.
Radiol 30:074–081. J Surg Oncol 94:572–586.
Hilgard, P. et al. (2010). Radioembolization with Kuo, J.C. et al. (2014). Serious hepatic
yttrium-90 glass microspheres in hepatocel- complications of selective internal radiation
lular carcinoma: European experience on therapy with yttrium-90 microsphere radio-
safety and long-term survival. Hepatology embolization for unresectable liver tumors.
52:1741–1749. Asia-Pac J Clin Oncol 10:266–272.
Jakobs, T.F. et al. (2008). Fibrosis, portal Lam, M.G.E.H., Banerjee, A., Louie, J.D., Sze, D.Y.
hypertension, and hepatic volume changes (2013a). Splenomegaly-associated throm-
induced by intra-arterial radiotherapy bocytopenia after hepatic yttrium-90 radio-
with 90yttrium microspheres. Dig Dis Sci embolization. Cardiovasc Intervent Radiol
53:2556–2563. 37:1009–1017.
Kao, Y.H. (2014). Non-target activity detection Lam, M.G.E.H. et al. (2013b). Safety of repeated
by post-radioembolization yttrium-90 PET/ yttrium-90 radioembolization. Cardiovasc
CT: Image assessment technique and case Intervent Radiol 36:1320–1328.
examples. Front Oncol 4: 11. Lam, M., Smits, M. (2013c). Value of 99mTc-
Kao, Y.H. et al. (2013). Post-radioembolization macroaggregated albumin SPECT for radio-
yttrium-90 PET/CT—Part 2: Dose-response embolization treatment planning. J Nucl
and tumor predictive dosimetry for resin Med 54:1681–1682. Available at: http://jnm.
microspheres. EJNMMI Res 3:1–1. snmjournals.org/cgi/content/full/54/9/1681.
Kennedy, A. et al. (2007). Recommendations Lencioni, R., Llovet, J. (2010). Modified RECIST
for radioembolization of hepatic malig- (mRECIST) assessment for hepatocellular car-
nancies using yttrium-90 microsphere cinoma. Semin Liver Dis 30:052–060.
302 The use of postprocedural imaging in the medical management of patients

Lhommel, R. et al. (2009). Yttrium-90 TOF PET Riaz, A. (2014). Side effects of yttrium-90 radio-
scan demonstrates high-resolution biodistri- embolization. Front Oncol 4:198.
bution after liver SIRT. Eur J Nucl Med Mol Sacks, A. et al. (2011). Value of PET/CT in the
Imaging 36:1696. management of liver metastases, Part 1. Am J
Mascarenhas, N., Ryu, R., Salem, R. (2011). Roentgenol 197:W256–W259.
Hepatic radioembolization complicated by Sag, A.A., Savin, M.A., Lal, N.R., Mehta, R.R.
abscess. Semin Intervent Radiol 28:222–225. (2014). Yttrium-90 radioembolization of malig-
Miller, F.H. et al. (2007). Response of liver metas- nant tumors of the liver: Gallbladder effects.
tases after treatment with yttrium-90 micro- Am J Roentgenol 202:1130–1135.
spheres: Role of size, necrosis, and PET. AJR Salem, R. et al. (2002). Yttrium-90 microspheres:
Am J Roentgenol 188:776–783. Radiation therapy for unresectable liver can-
Miller, A.B., Hoogstraten, B., Staquet, M., cer. J Vasc Interv Radiol 13:S223–S229.
Winkler, A. (1981). Reporting results of cancer Salem, R. et al. (2008). Incidence of radiation
treatment. Cancer 47:207–214. pneumonitis after hepatic intra-arterial
Minocha, J., Lewandowski, R. (2011). radiotherapy with yttrium-90 microspheres
Radioembolization for hepatocellular carci- assuming uniform lung distribution. Am J Clin
noma complicated by biliary stricture. Semin Oncol 31:431–438.
Intervent Radiol 28:226–229. Salem, R. et al. (2010). Radioembolization for hepa-
Neperud, J. (2013). Can imaging patterns of tocellular carcinoma using yttrium-90 micro-
neuroendocrine hepatic metastases predict spheres: A comprehensive report of long-term
response yttruim-90 radioembolotherapy? outcomes. Gastroenterology 138:52–64.
WJR 5:241. Salem, M.E. et al. (2013a). Radiographic param-
Northover, J.M., Terblanche, J. (1979). A new eters in predicting outcome of patients
look at the arterial supply of the bile duct in with hepatocellular carcinoma treated with
man and its surgical implications. Br J Surg yttrium-90 microsphere radioembolization.
66:379–384. ISRN Oncol 2013:1–8.
Pasciak, A.S. et al. (2015). The impact of an antire- Salem, R., Mazzaferro, V., Sangro, B. (2013b).
flux catheter on target volume particulate dis- Yttrium 90 radioembolization for the treat-
tribution in liver-directed embolotherapy: A ment of hepatocellular carcinoma: Biological
pilot study. J Vasc Interv Radiol. 26:660–669. lessons, current challenges, and clinical per-
Piduru, S.M. et al. (2012). Prognostic value of spectives. Hepatology 58:2188–2197.
18f-fluorodeoxyglucose positron emission Salem, R., Thurston, K.G. (2006).
tomography-computed tomography in pre- Radioembolization with 90yttrium micro-
dicting survival in patients with unresectable spheres: A state-of-the-art brachytherapy
metastatic melanoma to the liver undergoing treatment for primary and secondary
yttrium-90 radioembolization. J Vasc Interv liver malignancies. J Vasc Interven Radiol
Radiol 23:943–948. 17:1251–1278.
Poon, R.T.-P., Fan, S.T., Tsang, F.H.-F., Wong, J. Sangro, B. et al. (2006). Radioembolization using
(2002). Locoregional therapies for hepatocel- 90Y-resin microspheres for patients with
lular carcinoma: A critical review from the sur- advanced hepatocellular carcinoma. Radiat
geon’s perspective. Ann Surg 235:466–486. Oncol Biol 66:792–800.
Reiner, C.S. et al. (2014). Early treatment Sangro, B. et al. (2008). Liver disease induced
response evaluation after yttrium-90 radioem- by radioembolization of liver tumors:
bolization of liver malignancy with CT perfu- Description and possible risk factors. Cancer
sion. J Vasc Interv Radiol 25:747–759. 112:1538–1546.
Rhee, T.K. et al. (2008). Tumor response after Sato, K. et al. (2006). Treatment of unre-
yttrium-90 radioembolization for hepatocel- sectable primary and metastatic liver
lular carcinoma: Comparison of diffusion- cancer with yttrium-90 microspheres
weighted functional MR imaging with (TheraSphere®): Assessment of hepatic
anatomic MR imaging. J Vasc Interv Radiol arterial embolization. Cardiovasc Intervent
19:1180–1186. Radiol 29:522–529.
References 303

Schlaak, J. (2013). Choi criteria are superior in European Organization for Research and
evaluating tumor response in patients treated Treatment of Cancer, National Cancer Institute
with transarterial radioembolization for hepa- of the United States, National Cancer Institute of
tocellular carcinoma. Oncol Lett 6:1707–1712. Canada. J Natl Cancer Inst 92:205–216.
Shim, J.H. et al. (2012). Which response criteria Veloso, N. et al. (2013). Gastroduodenal ulcer-
best help predict survival of patients with ation following liver radioembolization with
hepatocellular carcinoma following chemoem- yttrium-90. Endoscopy 45:E108–E109.
bolization? A validation study of old and new Vouche, M. et al. (2015). Clinical imaging. J Clin
models. Radiology 262:708–718. Imaging 39:454–462.
Singh, P., Anil, G. (2013). Yttrium-90 radioemboli- Wang, L.M., Jani, A.R., Hill, E.J., Sharma, R.A.
zation of liver tumors: What do the images tell (2013). Anatomical basis and histopathologi-
us? Cancer Imaging 13:645–657. cal changes resulting from selective internal
Smith, M.T., Johnson, D.T., Gipson, M.G. (2015). radiotherapy for liver metastases. J Clin
Skin necrosis resulting from nontarget Pathol 66:205–211.
embolization of the falciform artery dur- Willowson, K., Tapner, M., Bailey, D. (2015).
ing transarterial chemoembolization with A multicentre comparison of quantitative
drug-eluting beads. Semin Intervent Radiol 90Y PET/CT for dosimetric purposes after
32:22–25. radioembolization with resin microspheres.
Smits, M.L.J. et al. (2013). Clinical and laboratory Eur J Nucl Med Mol Imaging:1–21.
toxicity after intra-arterial radioemboliza- Wondergem, M. et al. (2013). 99mTc-mac-
tion with 90Y-microspheres for unresectable roaggregated albumin poorly predicts
liver metastases Aravindan N, ed. PLoS One the intrahepatic distribution of 90Y resin
8:e69448. microspheres in hepatic radioemboliza-
Soydal, C. et al. (2013). The prognostic value of tion. J Nucl Med 54:1294–1301. Available at:
quantitative parameters of 18F-FDG PET/CT http://jnm.snmjournals.org/cgi/doi/10.2967/
in the evaluation of response to internal radia- jnumed.112.117614.
tion therapy with yttrium-90 in patients with Wright, C.L. et al. (2012). Radiation pneumonitis
liver metastases of colorectal cancer. Nucl following yttrium-90 radioembolization: Case
Med Commun 34:501–506. report and literature review. J Vasc Interv
Strigari, L. et al. (2010). Efficacy and toxicity Radiol 23:669–674.
related to treatment of hepatocellular car- Yu, J.-S. et al. (2002). Predisposing factors of
cinoma with 90Y-SIR spheres: Radiobiologic bile duct injury after transcatheter arte-
considerations. J Nucl Med 51:1377–1385. rial chemoembolization (TACE) for hepatic
Therasse, P. et al. (2000). New guidelines to evaluate malignancy. Cardiovasc Intervent Radiol
the response to treatment in solid tumors. 25:270–274.
PART 5
New Horizons

15 Future directions in radioembolization 307


Alexander S. Pasciak, J. Mark McKinney, and Yong C. Bradley
15
Future directions in radioembolization

ALEXANDER S. PASCIAK, J. MARK MCKINNEY, AND YONG C. BRADLEY

15.1 Introduction 307 15.5 Toward improved posttreatment


15.2 Background 308 imaging 315
15.3 Toward improved treatment 15.5.1 New tracers and methods 315
planning 309 15.5.2 Image analysis 316
15.4 Improving efficacy and safety 311 15.6 Toward extrahepatic tumor
15.4.1 Enhancement of T:N 311 radioembolization 318
15.4.2 Alternative methods to 15.7 Conclusions 318
enhance tumor targeting 314 References 319

normally have over patients undergoing radio-


15.1 INtrODUCtION embolization therapy have largely driven the
radioembolization technical process in the
From the perspective of medical physicists, past. Evidence for this can be seen in Chapters
nuclear medicine radiologists, and radiation 4 and  5, where the manufacturer-recommended
oncologists involved in radioembolization, the treatment-planning methods for radioemboliza-
ideal direction of the future of this therapy may tion are largely based on simple empiric calcula-
appear different than it does to interventional tions that are only patient specific at the simplest
radiologists. This is primarily due to the differ- level. For example, recall the specifics of the
ences in the common procedures in which each of body surface area (BSA) model often employed
these groups normally participate. Interventional for treatment planning using resin microspheres.
radiologists focus on procedures that, while tech- In this model, the impact of height and weight
nically demanding, normally do not involve the on the prescribed dosage exceed the impact of
physics, mathematics, or the related precision relative tumor burden. Additionally, the tumor
commonly employed in radiation oncology and type, vascularity, prior treatment, homogeneity
nuclear medicine. Further, while the technical of uptake, and any other patient-specific factors
considerations involved in a complex vascular are not considered. While BSA is widely used in
intervention necessitate careful review of pre- medicine for determining dosages for medica-
treatment structural and angiographic imaging, tions such as chemotherapy, it certainly makes
most nuclear medicine procedures are com- more sense for a systemic administration than
pletely imaging based, focusing on both function for a local brachytherapy such as radioemboli-
and structure to diagnose and treat the patient. zation. It is for these reasons that many medical
The distinction between these two groups and physicists, nuclear medicine radiologists, and
the ownership that interventional radiologists radiation oncologists new to radioembolization

307
308 Future directions in radioembolization

find themselves immediately frustrated with the ● Determining delivered dose (medical physicist,
seeming lack of precision associated with the cur- technologist)
rent standard of care in radioembolization treat- ● Posttreatment yttrium-90 (90Y) imaging
ment planning. It should be noted that this brief (nuclear medicine radiologist, medical physi-
discussion of the commonly employed BSA treat- cist, technologist)
ment planning method for resin microspheres is ● Surveying and clearing radioactive contamina-
not unique in its lack of tumor specificity—the tion (technologist, health physicist)
recommended method for glass microspheres is ● Releasing the patient and providing release
even less tumor specific and does not account for instructions (health physicist, medical physi-
tumor burden at all. Those looking for a more cist, technologist)
detailed review of these methods should refer to ● Using posttreatment 90Y imaging to plan future
Chapter 5. treatments (interventional radiologist, nuclear
While these treatment-planning methodologies medicine radiologist, radiation oncologist,
may be simplistic, radioembolization is still success- medical physicist)
ful and beneficial for many patients. Nevertheless,
improving the treatment-planning process for both In this chapter, we will discuss how the future
glass and resin microspheres is one way that the of radioembolization will be affected by improve-
future of radioembolization may be shaped. As the ments related to some of the above tasks. The
subject of this text would suggest, radioemboliza- authors acknowledge that radioembolization is a
tion is an inherently multidisciplinary field requir- field that has grown tremendously in recent years
ing not only an interventional radiologist but also but is still relatively young compared with alterna-
the skills of many other medical specialties. There tive treatments for liver cancer.
are numerous necessary steps required to perform
a safe and effective radioembolization and with so
many potential hands involved, the task can some-
times seem overwhelming. While covered in some 15.2 BaCKGrOUND
detail in the previous chapters, a summary list of
tasks required and potential specialties involved The use of 90Y and other radionuclides with local-
are shown below, with a particular focus on the ized energy deposition in the percutaneous treat-
tasks necessary for the audience of this text. ment of disease has a more lengthy history than
one might initially suspect. Before the widespread
● Patient recruitment and selection (interven- use of hepatic radioembolization, simpler percu-
tional radiologist, oncologist) taneous procedures were being performed clini-
● Vascular treatment planning (interventional cally for patients with chronic synovitis due, in
radiologist) part, to hypertrophy of the synovial membrane.
● Evaluating lung shunt fraction (interventional Ansell et  al. (1963) used a gold-198 (198Au) col-
radiologist, nuclear medicine radiologist) loid percutaneously injected into the synovial
● Dosimetric treatment planning (nuclear medi- space to destroy the superficial luminal layers of
cine radiologist, radiation oncologist, medical the synovial membrane. However, as this form of
physicist, interventional radiologist) therapy expanded, 90Y in various chemical forms
● Safely preparing the radioembolization dosage soon replaced 198Au (Oka et al., 1971; Prosser et
(technologist, radiopharmacy, medical physi- al., 1993; Stucki et al., 1993; Asavatanabodee et
cist, health physicist) al., 1997; Jahangier et al., 1997; Taylor et al., 1997;
● Preparing the angiographic suite (technologist, Jacob et al., 2003; Oztürk et al., 2008; Thomas
health physicist) et al., 2011).
● Controlling entry and exit into the treatment Radiation synovectomy using 90Y is a treat-
room (technologist, health physicist, medical ment that has seen some clinical use, particu-
physicist) larly for chronic synovitis that is refractory to
● Delivering the dosage (interventional traditional intra-articular steroid injections.
radiologist) Treatment traditionally has been used for
15.3 Toward improved treatment planning 309

rheumatoid arthritis, osteoarthritis (Taylor et al., intra-arterial infusion of resin 90Y microspheres
1997), psoriatic arthritis (Stucki et al., 1993), and with chemotherapy in the form of 5-fluoroura-
hemophilic arthritis (Thomas et al., 2011) unre- cil. Ariel’s patients received relatively large dos-
sponsive to systemic medical therapy. 5  mCi of ages of 90Y, ranging from 100 to 150 mCi (3.7–5.5
90Y silicate or 90Y resin-colloid injected into the GBq) that led to improved response in their 65
knee is capable of producing an absorbed dose in patient cohort. Interestingly, Ariel infused 90Y
the synovium at a depth of 1 mm exceeding 50 microspheres into these patients using both per-
Gy (Oka et al., 1971). The goal of radiation syno- cutaneous delivery and delivery through open
vectomy is to create fibrosis in the hypertrophic laparotomy with a catheter inserted directly into
areas of the synovium. 90Y has been used exten- the hepatic artery. Since this initial experience,
sively for knees; however, for smaller joints, that the techniques and sophistication involved in
is, elbows and shoulders, 186Re is preferred due the manufacture and treatment with 90Y micro-
to the lower beta energy (Kavakli et al., 2008). spheres have tremendously improved.
However, the downside of 186Re is the gamma The therapeutic percutaneous uses of endovas-
component of the decay, which could result in cular brachytherapy and radioembolization using
radiation dose to sensitive tissues near the injec- 90Y and other radionuclides have lengthy and inter-

tion site (e.g., lymph nodes). esting histories. However, this chapter will look to
Moving in the direction of endovascular the future of radioembolization.
therapy, vascular disease, one of the most com-
mon diseases in the world, has also been treated
with internal emitters. Percutaneous translumi-
15.3 tOWarD IMPrOVED
nal angioplasty (PTA) is one of the most com-
mon treatments for vascular stenosis. However,
trEatMENt PLaNNING
the durability of PTA is largely determined by
restenosis rates, which can be high. Prophylactic Treatment planning in 90Y radioembolization
endovascular brachytherapy (EVBT), as a preven- lacks much of the detail and patient specificity
tative tool for restenosis due to intimal hyperpla- required for external beam radiation therapy.
sia (Amols, 1999), has been used successfully for This is logical since there is an element of con-
a number of years with various radionuclides and trol in external beam radiation therapy and even
in various parts of the body (Minar, 2012). While in conventional brachytherapy that is lacking
early uses of EVBT were based on iridium-192 in radioembolization—namely, physical flow
(192Ir), a low-energy gamma emitter (Schopohl dynamics that determine the final location of the
et al., 1996; Reynaert et al., 2001; Piermattei microspheres. While this process cannot be con-
et al., 2002), some newer techniques use high- trolled, it can be predicted to a limited extent.
energy beta particles from phosphorus-32 (32P) As discussed extensively in Chapters 4 and 5,
(Piermattei et al., 2003), rhenium-188 (188Re) technetium-99m (99mTc)-macroaggregated albu-
(Werner et al., 2012), strontium-90 (90Sr), or 90Y min (MAA) single-photon emission computed
(Coucke, 2009). The advantage of pure beta emit- tomography/computed tomography (SPECT/CT)
ters is, of course, the markedly reduced radiation can be used as a standard component of treatment
safety concerns associated with the procedure. planning using the partition model. Many authors
However, despite clinical efficacy, the technical have examined the validity of MAA as a radio-
difficulty of EVBT has hindered its widespread embolization surrogate, with no clear consensus
clinical use in favor of alternatives such as drug- (Knesaurek et al., 2010; Kao et al., 2012; Lam and
eluting stents for the management of restenosis. Smits, 2013; Lam et al., 2013; Wondergem et al.,
Radioembolization also has a more lengthy 2013; Garin et al., 2014; Lam and Sze, 2014) as to
clinical history than one might initially expect. its accuracy in the modeling of hepatic distribu-
Ariel and Padula (1978a, 1978b) in 1978 reported tion. The position of the catheter tip during both
the first cases of the clinical use of 90Y micro- infusion of MAA and radioembolization is among
spheres in the intra-arterial treatment of colorec- the most critical factors to the prognostic utility of
tal metastases to the liver. Ariel combined tumor to normal uptake ratio (T:N) measurements
310 Future directions in radioembolization

made from 99mTc-MAA SPECT/CT. Positioning of 99m Tc-MAA as a tool for prognostication of gas-
the catheter tip becomes especially critical when troduodenal NTE (Sabet et al., 2011).
it is near a bifurcation or when it is positioned in Stepping away from the standard of care 99mTc-
a tortuous vessel (Jiang et al., 2012; Wondergem MAA for evaluating lung shunt fraction and treat-
et al., 2013). However, in spite of variable correla- ment simulation, there have been several attempts
tion between MAA and 90Y microspheres in the to identify alternative tracers that can be applied to
literature, many authors agree that MAA is an this purpose. Mathias and Green (2008) described
excellent option for treatment planning and pre- a simple method for the production of gallium-68
dictive dosimetry. This has been discussed in detail (68Ga) MAA from the elutant of a conventional
in Chapters 4 through 6. germanium-68/68Ga generator. The size range of
The use of 99mTc-MAA as a treatment-planning MAA did not change with the binding of 68Ga and
guide certainly has potential utility in the prog- binding efficiency exceeded 99% (Mathias and
nostication of lung shunt fraction, intrahepatic Green, 2008). 68Ga is a convenient positron emis-
dose distribution, and presence of gastroduodenal sion tomography (PET) radiotracer that provides
nontarget embolization (NTE). Due to a handful fully quantitative imaging with a superior resolu-
of publications describing the difficulty of man- tion to 99mTc SPECT. 68Ga MAA may have utility in
aging patients with ulcerations from gastroduo- conventional pulmonary perfusion studies, evalu-
denal NTE, the majority of radioembolization ation of lung shunt fraction for radioembolization,
treatment centers carefully examine post-MAA and, potentially, improved intrahepatic treatment
nuclear imaging to look for the presence of NTE. planning given the improved resolution and quan-
However, the majority of treatment centers per- tification of PET/CT.
form only planar imaging of 99mTc-MAA, as this It is likely, however, that the largest source
is the most common method used to determine of error in the use of 99mTc-MAA as a 90Y micro-
the lung shunt fraction and is recommended in sphere surrogate is in particulate deposition dif-
the package insert for both resin and glass 90Y ferences rather than SPECT resolution limits.
microspheres. Many institutions can improve Therefore, the greatest advances in pretreatment
both the sensitivity and specificity of 99mTc- simulation may come from replacing MAA with
MAA for the determination of NTE with sev- a superior particle, rather than identification of a
eral simple protocol modifications. As discussed radionuclide with superior imaging characteristics
in Chapter 14, there is a substantial increase in to 99mTc. Biodegradable human serum albumin
the prognostic utility of MAA as a predictor for (HSA) microspheres have been proposed for use
extrahepatic NTE with the use of SPECT/CT in pulmonary perfusion studies labeled with 99mTc
compared with planar imaging. A detailed study or 86Y. HSA microspheres have also been proposed
by Ahmadzadehfar et  al. (2010) suggested that as an alternative form of radioembolization when
the relative sensitivity of detecting extrahepatic labeled with 188Re or 90Y (Schiller et al., 2008). A
NTE increased from 32% to 100% when SPECT/ potential benefit of HSA microspheres over MAA
CT was used compared with planar imaging. That is that the size and surface characteristics of the
said, however, an increase in sensitivity may lead microsphere much more closely approximate
to exclusion of patients from treatment owing to those of 90Y microspheres, as shown in Figure 1 of
false positives, such as free technetium. In Chapter Schiller et al. (2008). Naturally, the specific gravity
4, there is a detailed discussion of the biological of an HSA microsphere will also closely approxi-
half-life of MAA, binding efficiency, and effects of mate that of 90Y resin microspheres. As a tool for
free 99mTc. Free 99mTc in the form of pertechnetate pretreatment simulation, the biodegradability of
(99mTcO4 –) will show a strong uptake in the gas- HSA microspheres has a favorable characteristic
tric mucosa, potentially leading to false positives since it may be degraded and removed from the
when MAA is used to assess NTE. Standard of hepatic vasculature prior to 90Y radioembolization.
care prophylaxis should include oral administra- Currently, there are multiple clinical tri-
tion of sodium perchlorate (NaClO4) as a blocking als underway to evaluate the utility of alterna-
agent to prevent the uptake of 99mTcO4 – in gastro- tive microspheres for pretreatment planning,
duodenal tissues. Sodium perchlorate has been both in biodegradable and permanent physi-
shown to substantially increase the specificity of cal form. Potential imaging modalities for these
15.4 Improving efficacy and safety / 15.4.1 Enhancement of T:N 311

microspheres primarily include SPECT or PET,


but in some cases have extended to magnetic 15.4 IMPrOVING EFFICaCY
resonance imaging (MRI). Holmium-166 (166Ho) aND SaFEtY
microspheres (Quiremspheres, Quirem Medical,
The Netherlands) are relatively unique in radio- 15.4.1 ENHANCEMENT OF T:N
embolization for their multimodality imaging
and therapy potential. 166Ho decays with the emis- As discussed in detail in Chapters 5 and 8, the suc-
sion of a two medium-energy beta particles at a cess of radioembolization depends on delivering
high yield with average energies of 654 and 691 sufficient dosage to the tumor to elicit a therapeu-
keV. 166Ho also emits an 80 keV gamma ray with a tic response while sparing uninvolved liver tis-
lower yield (approximately 6.5 per 100 transforma- sue from excessive toxicity. In conventional lobar
tions) that can be effectively imaged using SPECT. therapy, it is the T:N that in many ways will define
Finally, Holmium is strongly paramagnetic, result- the balance between sufficient absorbed dose to the
ing in convenient visualization using MRI with tumor and limitation of dose to normal liver tissue.
high contrast and resolution. Examples of direct T:N is defined in Equation 15.1:
imaging of 166Ho by both MRI and SPECT are
shown in Figure 1.1, in Chapter 1. The clinical pro- A90 Y,tumor / Vtumor
T:N= (15.1)
tocol for 166Ho radioembolization takes advantage A90 Y,normal / Vnormal
of multimodality imaging and includes pretreat-
ment infusion of a low-activity dosage of 166Ho where, A90 Y,tumor is the 90Y activity (MBq) deposited
radioembolization as a tool for treatment planning in the tumor and A90 Y,normal is the activity depos-
and evaluation of lung shunt fraction via nuclear ited in uninvolved liver tissue. Vnormal and Vtumor
imaging. This process eliminates MAA altogether are the respective volumes of each. Tumor type,
in the 166Ho radioembolization treatment process. size, burden, prior treatments, and other patient-
Many current and previous investigations have specific physiological factors significantly affect
focused on improving pretreatment imaging and T:N, leading to a range of clinical values which
simulation for radioembolization in an effort can vary from nearly 15:1 to less than 1. Table 5.2
to improve treatment planning. However, while in Chapter 5 lists some typical T:N values from
these tools can help to predict tumor and normal the literature. While there are many factors in
liver tissue absorbed doses, this is just part of the addition to T:N that determine the success or fail-
treatment-planning equation. Patient-specific ure of a radioembolic therapy, lower T:Ns make a
treatment planning also requires a precise knowl- robust clinical response difficult to achieve. The
edge of tumoricidal dose thresholds, which will premise behind the difficulty of successful radio-
vary at a minimum on tumor type, size, vascu- embolization in the setting of a low T:N (<2:1)
larity, previous therapy, and use of glass or resin can be linked back to the fundamental fallacy of
microspheres. The difficulty of obtaining these treating liver cancer with external beam radiation
data has been compounded in the past by the lim- therapy: the radiation toxicity threshold for unin-
ited availability of postradioembolization quan- volved liver tissue is low—potentially lower than
titative imaging; however, some excellent sources the absorbed dose necessary for effective tumor
do exist particularly for hepatocellular carcinoma control.
(HCC) (Strigari et al., 2010). Going forward, the Several methods of prophylactically increas-
discovery of 90Y PET/CT will open up the door to ing T:N are available and may improve efficacy in
precisely defining toxicity thresholds for radioem- certain classes of patients receiving lobar therapy.
bolization. An international clinical trial spon- Patients with low T:N (<2:1), or patients with mod-
sored by SIRTeX medical (SIR-Spheres®, SIRTex erate T:N (<3:1) in the setting of underlying liver
Technology Pty, Lane Cove, NSW, Australia) will disease and/or tumors that require large absorbed
use postradioembolization 90Y PET/CT to eluci- doses, may benefit from these techniques. Both
date dose–response thresholds based on tumor pharmaceutical and physical techniques can be
type for metastatic breast and colon cancer. Some used to prophylactically increase T:N prior to
initial data from this effort have already been pub- radioembolization in patients who fall into either
lished (Willowson et al., 2015). of these categories.
312 Future directions in radioembolization

15.4.1.1 Pharmaceutical methods muscle coat (Mattsson et al., 1977; Hafström et al.,
1980; Burke et al., 2001). A recent review by van
Tumor angiogenesis results in the formation of den Hoven et al. (2014b) summarized the results of
arterioles and capillaries that are structurally and all published studies that quantified the degree of
physiologically abnormal and support modifica- change in T:N before and after infusion of angio-
tion of T:N using both pharmaceutical and physi- tensin II. In these publications spanning findings
cal techniques. Tumor arterioles are abnormal in 71  patients, an increase in T:N ranging from
in that they do not have complete formation of 180% to 310% following infusion of angiotensin II
smooth muscle coat and lack autonomic inner- has been reported. These changes, which are sub-
vation and, therefore, autoregulatory response stantial, can dramatically affect the distribution of
(Mattsson et al., 1977; Ashraf et al., 1996; Burke radioactive microspheres. Figure 15.1 shows the
et al., 2001; Tanaka et al., 2008). Tumor capillaries absorbed dose to the tumor and to the uninvolved
are abnormal, owing to both incomplete formation liver as a function of T:N with the pre- and postan-
of the basement membrane and pericyte detach- giotensin II (van den Hoven et al., 2014b) ranges,
ment (Nagy et al., 2009). highlighted to illustrate the potential benefit.
Intra-arterial infusion of the vasoconstrictor Angiotensin II is not the only vasoconstric-
angiotensin II has been used in several studies tor that has been evaluated as a tool to improve
(Sasaki et al., 1985; Goldberg et al., 1991a, 1991b; liver-directed therapies. The effect of intra-arterial
Burke et al., 2001; Flower et al., 2001) as a means infusion of antidiuretic hormone (ADH) has
to preferentially constrict arterioles feeding nor- been demonstrated in a porcine model (Durack
mal liver tissue, while arterioles supplying tumor et al., 2012). Specifically, the benefits of ADH have
will remain largely unaffected owing, again, to been quantified in terms of its ability to reduce
the lack of innervation and incomplete smooth NTE by preferential constriction of gastroenteric

4
No angiotensin II, 2:1 T:N, 150 Gy tumor dose
Angiotensin II lower-limit effect, 150 Gy tumor dose
3.5 Angiotensin II upper- limit effect, 150 Gy tumor dose

2.5
Dosage (GBq)

1.5

0.5

0
0 10 20 30 40 50 60 70 80
Percentage tumor burden (%)

Figure 15.1 The theoretical treatment dosage (GBq) of yttrium-90 (90Y) radioembolization neces-
sary to achieve an average tumor absorbed dose of 150 Gy. Baseline in the absence of intra-arterial
antiotensin II administration assumes a 2:1 tumor to normal uptake ratio (T:N) and a 1300 cc total liver
volume. The slope of the curve indicates the necessary increase in the ideal treatment dosage as a
function of percentage tumor infiltration. With angiotensin II and its related lower and upper limit
of efficacy on T:N (van den Hoven et al., 2014b), the treatment dosage required to achieve 150 Gy is
decreased substantially at low tumor infiltration. As percentage tumor infiltration rises, the benefit of
angiotensin II is reduced. (From van den Hoven, A.F. et al., PLoS ONE, 9, e86394, 2014b.)
15.4 Improving efficacy and safety / 15.4.1 Enhancement of T:N 313

collaterals. ADH was effective in reducing the radioembolization to prevent extrahepatic NTE.
ratio of gastric to hepatic activity by a factor of two The use of specialty catheters, or antireflux cath-
(Durack et al., 2012). Similar analyses based on eters, such as the Surefire Infusion System (Surefire
infusion of catecholamines (Hafström et al., 1980; Medical Inc., Westminster, CO), may also protect
Tanaka et al., 2008) have been performed on both extrahepatic tissues from NTE by preventing ret-
rats (Hafström et al., 1980) and humans (Tanaka rograde flow (Arepally et al., 2013; Fischman et al.,
et al., 2008) with concomitant increases in T:N 2014; van den Hoven et al., 2014a; Morshedi et al.,
from norepinephrine (Hafström et al., 1980) and 2015) of microspheres into unprotected collaterals.
epinephrine (Tanaka et al., 2008). While vasocon- However, antireflux catheters (Figure 15.2) have
strictors can illicit a positive increase on T:N that recently been shown to alter the hepatic distribu-
may contribute to the improved safety and efficacy tion of radioembolization and other liver-directed
of radioembolization in some patients, their use therapies (Arepally et al., 2013; Pasciak et al., 2015;
must be weighed against the potential contrain- van den Hoven et al., 2015). Arepally et al. (2013)
dications related to their systemic effects. Median showed increases in distal arterial penetration
increases in systolic blood pressure of more than of tantalum microspheres following renal artery
40 mm Hg (Sasaki et al., 1985; Goldberg et al., embolization in a porcine model with the use of
1991a) have been reported following hepatic arte- an antireflux microcatheter compared with a con-
rial infusion of angiotensin II, and while transient, ventional end-hole catheter, sparking interest in
this increase may be an important safety consider- the use of these devices for a purpose other than
ation in some patients. preventing reflux.
While pharmaceutical techniques for increas-
15.4.1.2 Physical techniques ing T:N take advantage of the lack of complete
smooth muscle coat and innervation in tumor
As described in Chapter 3, pretreatment occlusion arterioles, physical techniques build upon the
of the right gastric artery (RGA) and gastroduo- related inability of tumor arterioles to autoregu-
denal arteries (GDA) is often performed prior to late in the setting of changing arterial pressure.

Figure 15.2 The Surefire Precision Infusion system. (Surefire Medical, Denver, CO.)
314 Future directions in radioembolization

Rose et al. (2013) made in vivo downstream hepatic distribution of 99mTc-MAA with an end-
hepatic-arterial blood pressure measurements hole catheter and an antireflux catheter are shown
with the expandable tip (Figure 15.3) of an in Figure 15.4a and b, respectively. The distribu-
antireflux catheter open and with it closed. The tion of 90Y resin microspheres following infusion
findings of this study indicated that the open with the antireflux catheter and imaged directly
tip of the antireflux catheter reduced down- using 90Y PET/CT is shown in Figure  15.4d and
stream systolic and diastolic arterial pressure by demonstrates excellent agreement to Figure 15.4b.
nearly 50%. In theory, owing to the autoregula- While it is convenient to cite downstream
tory response of normal arteries and arterioles, pressure changes to explain the effect of an anti-
radioembolization performed using an antireflux reflux catheter, it is likely that centering of the
microcatheter may trigger vasoconstriction of tip as well as turbulence of flow also contributes
the vessels perfusing uninvolved hepatic paren- to the impact of these devices. For example, the
chyma. At the same time, the structurally abnor- open semiocclusive tip (Figure 15.3) will result
mal angiogenesis-induced tumor arterioles are in an increase in the turbulence of downstream
not likely to constrict owing to absence of smooth arterial flow. Increased turbulence of flow may
muscle, innervation, and autoregulatory proper- result in a more homogenous cross-sectional
ties (Mattsson et al., 1977; Burke et al., 2001). This distribution of microspheres in downstream
process could preferentially shunt microspheres arteries, potentially improving homogeneity of
toward the tumor compartment, temporarily deposition (van den Hoven et al., 2015).
increasing the T:N.
The effect of an antireflux catheter on T:N in
radioembolization has been shown experimen- 15.4.2 ALTERNATIVE METHODS
tally using 99mTc-MAA with a two-step same-day TO ENHANCE TUMOR
infusion (Figure 15.4) based on a protocol tradi- TARGETING
tionally used for routine renal and cardiac per-
fusion imaging (Pasciak et al., 2015). Statistically A more proactive approach to increasing T:N
significant increases in tumor uptake, commen- involves the occlusion of arteries supplying
surate with decreases in uninvolved liver, sup- uninvolved areas of the liver in an effort to redi-
port the premise that an antireflux catheter can rect hepatic-arterial blood flow, and thus 90Y
increase T:N (Pasciak et al., 2015). Differences in microspheres, to the tumor itself. Naturally,

(a) (b)

Figure 15.3 Proper hepatic angiogram showing (a) an end-hole catheter and (b) an antireflux catheter
with the tip expanded.
15.5 Toward improved posttreatment imaging / 15.5.1 New tracers and methods 315

(b)
(a) 15.5 tOWarD IMPrOVED
POSttrEatMENt IMaGING

15.5.1 NEW TRACERS AND


METHODS
In the past, the difficulty of directly imaging 90Y has
led to a variety of interesting experimental modifi-
cations to 90Y microspheres. Aliva-Rodriguez was
(c) (d)
able to successfully bind 86Y and 89Zr to the surface
of resin 90Y microspheres. Spheres were radiolabeled
and the binding stability for in vivo applications was
confirmed out to 24 hours at a physiological pH tem-
perature (Avila-Rodriguez et  al., 2007). This tech-
nique had the benefit of direct quantitative imaging
of 86Y and 89Zr using PET/CT. Investigations into
dual isotope 90Y and 177Lu microspheres have also
been performed (Poorbaygi et al., 2011), with effec-
tive imaging of the gamma emissions of 177Lu using
SPECT/CT. 166Ho radioembolization, as already dis-
cussed, is perhaps the best in multimodality direct
imaging of radioactive microspheres.
Figure 15.4 (a) Single-photon emission com- Chapters 10 and 11 discussed in detail tech-
puted tomography (SPECT) following infusion
niques for quantification of 90Y bremsstrahlung
of technetium-99m-macroaggregated albumin
(99mTc-MAA) using a conventional end-hole
SPECT and 90Y PET/CT, which have been substan-
catheter and (b) using an antireflux catheter. tially refined over the past 5 years. Particularly in
The catheter tip position was identical in each the case of 90Y PET/CT, many modern PET/CT sys-
case. (c) The distribution of 99mTc-MAA can be tems are able to accurately quantify 90Y right out of
compared with contrast-enhanced computed the box using existing software (Willowson et al.,
tomography (CT) in this patient with focal 2015). In light of this information, the driving force
hepatocellular carcinoma (HCC). (d) 90Y positron behind development of directly imageable micro-
emission tomography (PET)/CT following infusion spheres using alternative tracers has decreased
of resin 90Y microspheres with the antireflux cath-
substantially. This is not to say, however, that there
eter. The distribution of 90Y was well predicted
with 99mTc-MAA, as shown in (b).
are no areas for improvement.
As quantitative imaging of 90Y radioemboliza-
such efforts would only be indicated with tem- tion becomes more common, routine techniques
porary methods of occlusion such as biodegrad- normally reserved for diagnostic studies can be
able starch microspheres or gelatin powder. applied. One such example is respiratory gating,
Degradable starch microspheres (DSM) have which has been shown to affect both lesion size and
been shown in a porcine model to have a short quantification in 2-deoxy-2-[fluorine-18]fluoro-
biological half-life with complete re-perfusion d-glucose (18FDG) PET/CT (Suenaga et al., 2013).
in the liver in 30 minutes (Pieper et al., 2015). This technique can be applied to postradioembo-
DSM may have utility in improving safety by lization quantitative imaging, including 90Y PET/
protecting normal liver tissue in situations where CT (Pasciak et al., 2014), and may be capable of
subselection or repositioning of the microcath- improving lesion detection and quantitative accu-
eter to avoid microsphere deposition in large racy. Figure 15.5 visually illustrates the advantages
volumes of uninvolved liver is not a possibility of amplitude-based respiratory gating in postra-
(Meyer et al., 2013). dioembolization imaging.
316 Future directions in radioembolization

(a) (b)

(c)

Figure 15.5 (a) Posttreatment 90Y PET/CT on a Siemens mCT Flow without respiratory gating and
(b) using amplitude-based gating (Siemens HD•Chest). Figure parts (a) and (b) were both recon-
structed from the same acquisition data, making the decreased segment VIII lesion size easy to
appreciate when amplitude-based gating is used. Respiratory motion can effect image quality and
quantification in hepatic 90Y PET/CT just as it can in 2-deoxy-2-[fluorine-18]fluoro- d -glucose (18FDG)
PET/CT. (c) Pretreatment hepatic protocol magnetic resonance imaging (MRI) for comparison.

15.5.2 IMAGE ANALYSIS ● There are many ways to contour the tumor.
Contouring based on 90Y uptake on postradio-
The increasing use of quantitative postradioem- embolization imaging or contouring on pre-
bolization imaging based on either alternative treatment 18FDG PET/CT, contrast-enhanced
radiotracers, 90Y PET/CT, or quantitative brems- CT, or hepatic protocol MRI?
strahlung SPECT suggests the need for standard- ● Published dose–response thresholds are based
ized techniques to interpret these images. While on a particular method of dose characteriza-
there are numerous methods that can be used tion that may or may not be reproducible or
to convert quantitative 90Y images into three- even specified in the literature.
dimensional representations of absorbed dose ● Finally, the quantitative imaging modality
as described in Chapter 12, there is a significant used for postradioembolization imaging will
ambiguity in the simple process of using this infor- itself effect the dose measurement owing to
mation to determine the absorbed dose to a tumor. differences in quantification accuracy and
Using this interpreted tumor absorbed-dose data contrast recovery (a function of tumor size).
to predict treatment efficacy is associated with
several pitfalls: The future of radioembolization includes the
use of quantitative postradioembolization imag-
● What “dose metric” to the tumor correlates ing to predict treatment efficacy by comparison to
with published tumoricidal thresholds? tumoricidal thresholds. This is incredibly powerful
Average dose, maximum dose, something else? since it allows a physician to immediately consider
15.5 Toward improved posttreatment imaging / 15.5.2 Image analysis 317

alternative or adjuvant therapy, allowing radioem-


bolization to mesh synergistically with the spec-
trum of treatment options available to the patient.
Several examples in the literature have illustrated
the benefits of quantitative postradioembolization
imaging as a tool to influence patient care decisions
(Chang et al., 2013; Bourgeois et al., 2014; Pasciak
et al., 2014). These techniques are also discussed in
Chapter 14. (a) (b)
As we progress as a field we must become uni-
fied and consistent to avoid the aforementioned Figure 15.6 An example of count-preserving
pitfalls. To this end, the suggestions in Section deformable registration using the MIM 6 (MIM
15.5.2.1 may serve as a starting point. Software Inc., Cleveland, OH) software pack-
age. (a) Semiautomatic contouring of liver and
15.5.2.1 Tumor contouring on tumor is performed on hepatic protocol MRI.
(b) Deformable registration algorithm is used to
postradioembolization copy contours onto 90Y PET/CT posttreatment
quantitative imaging imaging. To preserve absorbed dose quantifica-
tion, contours are deformed to match the previ-
Posttreatment imaging of radioactive microspheres ously defined registration.
is not a diagnostic test for liver cancer and should
not be treated as such. Although contouring tumor
based on areas of high 90Y activity concentration diagnostic scan is referenced. Those involved
may sometimes correlate with active tumor, more in this task must understand that microsphere
often it will not. Therefore, this practice may lead to deposition is a mechanical process and, again,
unreliable correlation with dose–response thresh- postradioembolization imaging is not diagnostic
olds and poor prognostic accuracy of quantitative evaluation of tumor location or activity.
postradioembolization imaging. Instead, the fol-
lowing guidelines should be applied. 15.5.2.2 Dose metrics of interest
1. Tumor contours on quantitative posttreatment The most common dose metric reported in the lit-
imaging should be defined based on pretreat- erature is the average absorbed dose to the tumor
ment diagnostic scans. Pretreatment diagnostic (Davg). In many respects, Davg is flawed despite its
imaging may include 18FDG PET/CT, three- wide use for convenience. The failure of radioac-
phase hepatic CT, hepatic protocol MRI, and, tive microspheres to penetrate into small areas of
for example, specialty procedures such as 111In tumor will artificially reduce the Davg measure-
octreotide SPECT for neuroendocrine tumors. ments, even if the majority of the tumor may
Based on the patient’s history, the appropriate respond to the therapy. On the other hand, Davg
standard of care diagnostic imaging procedure may be artificially inflated if a small portion of the
should be selected by the care provider and tumor receives a large dose, even if the majority of
used to define tumor contours. it is left untreated.
2. Tumor contours on pretreatment imaging Alternatively, some authors have used stan-
should be translated onto postradioembo- dards from external beam radiation therapy such
lization quantitative imaging using count- as D70 and V100 for tumor analysis (Kao et al.,
preserving deformable image-registration 2013). D70 is the minimum absorbed dose deliv-
software. An example of this process is shown in ered to 70% of the tumor volume, while V100 is
Figure 15.6. the percentage of tumor volume exceeding 100
Gy. D70 is not skewed by either of the aforemen-
Certainly, the software in (2) above may not tioned scenarios and is a more reproducible met-
always be available. A qualified radiologist or ric for dosimetry based on postradioembolization
nuclear medicine physician can still reliably draw quantitative imaging. While not reported by
contours manually so long as the pretreatment some nuclear image analysis software packages,
318 Future directions in radioembolization

D70 can always be computed from dose–volume patient lived 23 months following RARE and
histogram data exported from a tumor contour. expired from extrarenal metastases; however, the
primary renal tumor remained well controlled
and stable (Hamoui et al., 2013). Several small-
15.5.2.3 Standardization scale clinical trials are underway to continue the
investigation into the utility of RARE for RCC.
Even if count-preserving deformable image regis-
Radioembolization of lung metastases via the
tration software is used to measure a robust dose
bronchial artery has also been performed in a small
metric such as D70, dose–response data used for
patient series by Ricke et al. (2013). Two patients,
comparison must also have been measured in a sim-
one with colorectal cancer and the another with
ilar reproducible way. Standardization as a field is
RCC lung metastasis who had failed chemotherapy
essential to utilizing dose–response data to influence
and were not surgical candidates, were treated using
the clinical decision-making process for a patient.
radioembolization via the bronchial artery (Ricke
In this regard, radioembolization is decades behind
et al., 2013). A conservative treatment plan was
external beam radiation therapy. Published guide-
assumed based on a T:N of 1:1 owing to the differ-
lines from the Society of Nuclear Medicine, Society
ing arterial anatomy in the lung and liver. 200 MBq
of Interventional Radiology, American Association
of 90Y resin microspheres was infused into both
of Physics in Medicine, or the American Society of
patients through a branch of the bronchial artery
Therapeutic Radiation Oncology would be helpful
perfusing multiple lung segments. The authors
in moving the field of radioembolization forward in
performed both posttreatment bremsstrahlung
the establishment of dose–response thresholds.
SPECT and 90Y PET/CT and found, surprisingly,
that pulmonary deposition of microspheres was
limited only to active tumor (Ricke et al., 2013). In
fact, the T:N for bronchial artery radioemboliza-
15.6 tOWarD EXtraHEPatIC
tion seems far higher than typical T:N for hepatic
tUMOr radioembolization. This finding is critical in that
raDIOEMBOLIZatION it supports future investigations into the utility of
radioembolization for lung metastases.
Several attempts at using radioembolization for A nononcologic extrahepatic use of radioembo-
extrahepatic tumor control and other endpoints lization has also recently been explored. Pasciak et
have been investigated. One that has received al. (2016) infused 90Y radioembolization into the
some attention is renal artery radioembolization gastric fundus in a porcine model to evaluate the
for renal cell carcinoma (RCC). While renal artery potential of radioembolization in the management
embolization with bland microspheres prior to of obesity. Although the animal cohort was small,
surgical resection is a widely used tool that aids decreased weight gain was noted. The mecha-
in the control of blood loss during surgery, renal nism of action was thought to be both a decrease
artery radioembolization as a treatment modal- in ghrelin producing cells in the gastric mucosa
ity is much more controversial. Over 20 years and a decrease in stomach size and volume. While
ago, selective renal artery radioembolization this is an interesting application of radioemboliza-
(RARE) using 90Y microspheres was performed tion, additional animal studies are needed before a
in a porcine model, with the capacity to deliver in human trial is considered.
excess of 100 Gy to the kidney with greater than
95% dose retention and sparing of extrarenal tis-
sues (Zimmermann et al., 1995). However, its use
in humans has never progressed beyond a few
15.7 CONCLUSIONS
case reports. One such example of a large (14.7 ×
11.1 cm) focal RCC mass was embolized with 90Y Many of the ideas provided in this chapter on the
glass microspheres delivering an average tumor future of radioembolization stand upon research
dose of 80 Gy (Hamoui et al., 2013). At 8 weeks done in the past. In some cases, interesting and novel
following RARE, CT imaging revealed patchy trials related to improving radioembolization were
tumor hypodensity consistent with necrosis. The performed years ago, before radioembolization was
References 319

widely used. However, the failure to widely adopt Ariel, I.M., Padula, G. (1978b). Treatment of
these methods may simply be due to the fact that symptomatic metastatic cancer to the liver
radioembolization has shown strong clinical effi- from primary colon and rectal cancer by the
cacy and safety as-is. This has made drastic changes intraarterial administration of chemotherapy
in the procedure or treatment difficult to justify. and radioactive isotopes. J Surg Oncol
That said, an improved understanding of radioem- 10:327–336.
bolization dose–response may be within reach due Asavatanabodee, P., Sholter, D., Davis, P. (1997).
to the availability of new imaging techniques such as Yttrium-90 radiochemical synovectomy in
90Y PET/CT. These data could have a broad impact chronic knee synovitis: A one year retrospec-
on both treatment planning, patient selection, and tive review of 133 treatment interventions.
patient follow-up. However, this information cannot J Rheumatol 24:639–642.
effectively be elucidated by individual researchers; Ashraf, S. et al. (1996). The absence of autonomic
instead, an international effort should be organized perivascular nerves in human colorectal liver
in order to obtain useful and reproducible results, metastases. Br J Cancer 73:349–359.
ideally through professional societies. Avila-Rodriguez, M.A. et al. (2007). Positron-
As more is understood about the biological emitting resin microspheres as surrogates of
effects of radioembolization, it will in turn will 90Y SIR-Spheres: A radiolabeling and stability
further the multidisciplinary nature of radio- study. Nucl Med Biol 34:585–590.
embolization, expanding the role of medical Bourgeois, A.C. et al. (2014). Intra-procedural
physicists, radiation oncologists, and nuclear 90Y PET/CT for treatment optimization of
medicine radiologists in this treatment modal- 90Y radioembolization. J Vasc Interv Radiol
ity. Because of the unique nature of this therapy, 25:271–275.
we believe this can only result in the improved Burke, D. et al. (2001). Continuous angiotensin
patient outcomes. II infusion increases tumour: Normal blood
flow ratio in colo-rectal liver metastases. Br J
REFERENCES Cancer 85:1640–1645.
Chang, T.T., Bourgeois, A.C., Balius, A.M.,
Ahmadzadehfar, H. et al. (2010). The signifi- Pasciak, A.S. (2013). Treatment modifica-
cance of 99mTc-MAA SPECT/CT liver per- tion of yttrium-90 radioembolization based
fusion imaging in treatment planning for on quantitative positron emission tomog-
90Y-microsphere selective internal radiation raphy/CT imaging. J Vasc Interv Radiol
treatment. J Nucl Med 51:1206–1212. 24:333–337.
Amols, H.I. (1999). Review of endovascular Coucke, P. (2009). Basic rules of dosimetry in
brachytherapy physics for prevention of reste- endovascular brachytherapy. J Interv Cardiol
nosis. Cardiovasc Radiat Med 1:64–71. 13:425–429.
Ansell, B.M., Crook, A., Mallard, J.R. (1963). Durack, J.C. et al. (2012). Intravenous vasopres-
Evaluation of intra-articular colloidal gold Au sin for the prevention of nontarget gastro-
198 in the treatment of persistent knee effu- intestinal embolization during liver-directed
sions. Ann Rheum Dis 22:435–439. cancer treatment: Experimental study
Arepally, A., Chomas, J., Kraitchman, D., Hong, in a porcine model. J Vasc Interv Radiol
K. (2013). Quantification and reduction of 23:1505–1512.
reflux during embolotherapy using an anti- Fischman, A.M. et al. (2014). Prospective, ran-
reflux catheter and tantalum microspheres: domized study of coil embolization versus
Ex vivo analysis. J Vasc Interv Radiol surefire infusion system during yttrium-90
24:575–580. radioembolization with resin microspheres.
Ariel, I.M., Padula, G. (1978a). Treatment of J Vasc Interv Radiol 25:1709–1716.
symptomatic metastatic cancer to the liver Flower, M.A. et al. (2001). 62Cu-PTSM and PET
from primary colon and rectal cancer by the used for the assessment of angiotensin
intra-arterial administration of chemotherapy II-induced blood flow changes in patients with
and radioactive isotopes. Prog Clin Cancer colorectal liver metastases. Eur J Nucl Med
7:247–254. 28:99–103.
320 Future directions in radioembolization

Garin, E., Boucher, E., Rolland, Y. (2014). 99mTc- Knesaurek, K. et al. (2010). Quantitative com-
MAA-based dosimetry for liver cancer treated parison of yttrium-90 (90Y)-microspheres and
using 90Y-loaded microspheres: Known proof technetium-99m (99mTc)-macroaggregated
of effectiveness. J Nucl Med 55:1391–1392. albumin SPECT images for planning 90Y
Goldberg, J.A. et al. (1991a). The use of angio- therapy of liver cancer. Technol Cancer Res
tensin II as a potential method of targeting Treatm 9:253–261.
cytotoxic microspheres in patients with intra- Lam, M.G.E.H. et al. (2013). Prognostic utility of
hepatic tumour. Br J Cancer 63:308–310. 90Y radioembolization dosimetry based on
Goldberg, J.A. et al. (1991b). Angiotensin II as a fusion 99mTc-macroaggregated albumin-
potential method of targeting cytotoxic-loaded 99mTc-sulfur colloid SPECT. J Nucl Med
microspheres in patients with colorectal liver 54:2055–2061.
metastases. Br J Cancer 64:114–119. Lam, M.G.E.H., Smits, M.L.J. (2013). Value of
Hafström, L., Nobin, A., Persson, B., Sundqvist, 99mTc-macroaggregated albumin SPECT for
K. (1980). Effects of catecholamines on car- radioembolization treatment planning. J Nucl
diovascular response and blood flow distribu- Med 54:1681–1682.
tion to normal tissue and liver tumors in rats. Lam, M.G.E.H., Sze, D.Y. (2014). Reply: 99mTc-
Cancer Res 40:481–485. MAA-based dosimetry for liver cancer treated
Hamoui, N. et al. (2013). Radioembolization of using 90Y-loaded microspheres: Known proof
renal cell carcinoma using yttrium-90 micro- of effectiveness. J Nucl Med 55:1392–1393.
spheres. J Vasc Interv Radiol 24:298–300. Mathias, C.J., Green, M.A. (2008). A convenient
Jacob, R., Smith, T., Prakasha, B., Joannides, route to [68Ga]Ga-MAA for use as a particu-
T. (2003). Yttrium90 synovectomy in the late PET perfusion tracer. Appl Radiat Isot
management of chronic knee arthritis: A 66:1910–1912.
single institution experience. Rheumatol Int Mattsson, J., Appelgren, L., Hamberger, B.,
23:216–220. Peterson, H.I. (1977). Adrenergic innerva-
Jahangier, Z.N., Jacobs, J.W., van Isselt, J.W., tion of tumour blood vessels. Cancer Lett
Bijlsma, J.W. (1997). Persistent synovitis 3:347–351.
treated with radiation synovectomy using Meyer, C. et al. (2013). Feasibility of temporary
yttrium-90: A retrospective evaluation of 83 protective embolization of normal liver tissue
procedures for 45 patients. Br J Rheumatol using degradable starch microspheres during
36:861–869. radioembolization of liver tumours. Eur J Nucl
Jiang, M., Fischman, A., Nowakowski, F.S. (2012). Med Mol Imaging 41:231–237.
Segmental perfusion differences on paired Minar, E. (2012). Commentary: Resuscitation
Tc-99m macroaggregated albumin (MAA) of endovascular brachytherapy owing
hepatic perfusion imaging and yttrium-90 to improved logistics. J Endovasc Ther
(Y-90) bremsstrahlung imaging studies in SIR- 19:476–479.
Sphere radioembolization: Associations with Morshedi, M.M., Bauman, M., Rose, S.C., Kikolski,
angiography. J Nucl Med Radiat Ther 3:122. S.G. (2015). Yttrium-90 resin microsphere
Kao, Y.H. et al. (2012). Image-guided personal- radioembolization using an antireflux cath-
ized predictive dosimetry by artery-specific eter: An alternative to traditional coil embo-
SPECT/CT partition modeling for safe and lization for nontarget protection. Cardiovasc
effective 90Y radioembolization. J Nucl Med Intervent Radiol 38:381–388.
53:559–566. Nagy, J.A., Chang, S.-H., Dvorak, A.M., Dvorak,
Kao, Y.H. et al. (2013) Post-radioembolization H.F. (2009). Why are tumour blood vessels
yttrium-90 PET/CT—Part 2: Dose-response abnormal and why is it important to know?
and tumor predictive dosimetry for resin Br J Cancer 100:865–869.
microspheres. EJNMMI Res 3:1–1. Oka, M., Rekonen, A., Ruotsi, A., Seppälä, O.
Kavakli, K. et al. (2008). Radioisotope syno- (1971). Intra-articular injection of Y-90 resin
vectomy with rhenium186 in haemophilic colloid in the treatment of rheumatoid
synovitis for elbows, ankles and shoulders. knee joint effusions. Acta Rheumatol Scand
Haemophilia 14:518–523. 17:148–160.
References 321

Oztürk, H., Oztemür, Z., Bulut, O. (2008). Sabet, A. et al. (2011). Significance of oral admin-
Treatment of skin necrosis after radiation istration of sodium perchlorate in planning
synovectomy with yttrium-90: A case report. liver-directed radioembolization. J Nucl Med
Rheumatol Int 28:1067–1068. 52:1063–1067.
Pasciak, A.S. et al. (2014). Radioembolization and Sasaki, Y. et al. (1985). Changes in distribution of
the dynamic role of (90)Y PET/CT. Front Oncol hepatic blood flow induced by intra-arterial
4:38. infusion of angiotensin II in human hepatic
Pasciak, A.S. et al. (2015). The impact of an anti- cancer. Cancer 55:311–316.
reflux catheter on target volume particulate Schiller, E. et al. (2008). Yttrium-86-labelled
distribution in liver-directed embolotherapy: human serum albumin microspheres: Relation
A pilot study. J Vasc Interv Radiol 26:660–669. of surface structure with in vivo stability. Nucl
Pasciak, A.S., Bourgeois, A.C., Paxton, B.E., Med Biol 35:227–232.
Nodit, L., Coan, P.N., Kraitchman, D., Stinnett, Schopohl, B. et al. (1996). 192Ir endovascular
S., Patel, V.M., Fu, Y., Adams, J.K., Tolbert, brachytherapy for avoidance of intimal hyper-
M.K., Lux, C.N., Arepally, A., Bradley, Y.C. plasia after percutaneous transluminal angio-
(2016). Bariatric radioembolization: A pilot plasty and stent implantation in peripheral
study on technical feasibility and safety in a vessels: 6 years of experience. Int J Radiat
porcine model. J Vasc Interv Radiol, in press. Oncol Biol Phys 36:835–840.
Pieper, C.C. et al. (2015). Temporary arte- Strigari, L. et al. (2010). Efficacy and toxicity related
rial embolization of liver parenchyma to treatment of hepatocellular carcinoma with
with degradable starch microspheres 90Y-SIR spheres: Radiobiologic considerations.
(EmboCept®S) in a swine model. Cardiovasc Journal of nuclear medicine: Official publica-
Intervent Radiol 38:435–441. tion. Soc Nucl Med 51:1377–1385.
Piermattei, A. et al. (2002). A standard dosim- Stucki, G. et al. (1993). Efficacy and safety of
etry procedure for 192Ir sources used for radiation synovectomy with yttrium-90:
endovascular brachytherapy. Phys Med Biol A retrospective long-term analysis of 164
47:4205–4221. applications in 82 patients. Br J Rheumatol
Piermattei, A. et al. (2003). Experimental 32:383–386.
dosimetry of a 32P catheter-based endovas- Suenaga, Y. et al. (2013). Respiratory-gated 18F-
cular brachytherapy source. Phys Med Biol FDG PET/CT for the diagnosis of liver metas-
48:2283–2296. tasis. Eur J Radiol 82:1696–1701.
Poorbaygi, H. et al. (2011). Applied radiation and Tanaka, O. et al. (2008). Epinephrine-infused CTHA
isotopes. Appl Radiat Isot 69:1407–1414. for HCCs. Abdom Imaging 33:308–312.
Prosser, J.S. et al. (1993). Induction of micronuclei Taylor, W.J., Corkill, M.M., Rajapaske, C.N. (1997)
in peripheral blood lymphocytes of patients A retrospective review of yttrium-90 synovec-
treated for rheumatoid or osteo-arthritis of the tomy in the treatment of knee arthritis. Br J
knee with dysprosium-165 hydroxide macroag- Rheumatol 36:1100–1105.
gregates or yttrium-90 silicate. Cytobios 73:7–15. Thomas, S. et al. (2011). Radioactive synovec-
Reynaert, N., Van Eijkeren, M., Taeymans, Y. tomy with Yttrium90 citrate in haemophilic
(2001) Dosimetry of 192Ir sources used for synovitis: Brazilian experience. Haemophilia
endovascular brachytherapy. Phys Med Biol 17:e211–e216.
46:499–516. van den Hoven, A.F. et al. (2014a). Posttreatment
Ricke, J., Großer, O., Amthauer, H. (2013). Y90 - PET-CT-confirmed intrahepatic radioemboliza-
radioembolization of lung metastases via tion performed without coil embolization, by
the bronchial artery: A report of 2 cases. using the antireflux Surefire Infusion System.
Cardiovasc Intervent Radiol 36:1664–1669. Cardiovasc Intervent Radiol 37:523–528.
Rose, S.C., Kikolski, S.G., Chomas, J.E. (2013). van den Hoven, A.F. et al. (2014b). The effect
Downstream hepatic arterial blood pres- of intra-arterial angiotensin II on the hepatic
sure changes caused by deployment of the tumor to non-tumor blood flow ratio for
surefire antireflux expandable tip. Cardiovasc radioembolization: A systematic review,
Intervent Radiol 36:1262–1269. Morishita R, ed. PLoS ONE 9:e86394.
322 Future directions in radioembolization

van den Hoven, A.F. et al. (2015). Innovation with resin microspheres: The QUEST
in catheter design for intra-arterial liver cancer Phantom Study. Eur J Nucl Med Mol Imaging
treatments results in favorable particle-fluid 42:1202–1222.
dynamics. J Exp Clin Cancer Res 34:74. Wondergem, M. et al. (2013). 99mTc-macroaggre-
Werner, M. et al. (2012). Endovascular brachy- gated albumin poorly predicts the intrahe-
therapy using liquid beta-emitting rhe- patic distribution of 90Y resin microspheres
nium-188 for the treatment of long-segment in hepatic radioembolization. J Nucl Med
femoropopliteal in-stent stenosis. J Endovasc 54:1294–1301.
Ther 19:467–475. Zimmermann, A. et al. (1995). Renal pathology
Willowson, K.P., Tapner, M.; QUEST Investigator after arterial yttrium-90 microsphere admin-
Team, Bailey, D.L. (2015). A multicentre istration in pigs. A model for superselective
comparison of quantitative (90)Y PET/CT for radioembolization therapy. Invest Radiol
dosimetric purposes after radioembolization 30:716–723.
Index

a Biliary sequelae, 60 Chemoembolization, 38, 40–41


Biliary stricture, 294 hepatocellular carcinoma, 43
Aberrant hepatic artery, 13
Biloma, 292–293 metastatic colorectal cancer, 44
Absolute activity calibration, 237
Biohazard bags, 162 Child–Pugh score, 34
Absorbed dose, 88–89, 201
Biological effective dose (BED), Choi criteria, 288–290
calculating, 93–94
173–174, 212 Cholangitis, 293–294
to different lobule structures, 206
Biological repopulation factor Chronic synovitis, 308
Acute pancreatitis, 295
(BRF), 174 Classic hepatic arterial supply, 54
ADH, see Antidiuretic hormone
Body surface area (BSA), 277, Clusters of microspheres, 4
Anatomical tumor response
307–308 Coaxial microcatheter technique, 57
assessment, 22
for resin microspheres, 98, 105 Coil embolization, 59
Angiogenesis, 61
Breast cancer (BrC), 4 Coil embolotherapy, 58
Angiotensin II, intra-
Bremsstrahlung single-photon Colorectal cancer (CRC), 4, 44–45,
arterial infusion of
emission computed 77
vasoconstrictor, 312
tomography with Colorectal carcinoma (CRC)
Antidiuretic hormone (ADH),
integrated computed metastasis, 283
312–313
tomography Common hepatic artery (CHA),
Antireflux catheter (ARC), 14, 18,
(Bremsstrahlung SPECT/ 12–13
53, 59, 61, 313, 314
CT), 264–266 Common terminology criteria for
AP, see Arterial perfusion
Bremsstrahlung x-rays, 254 adverse events (CTCAE),
Arterial access, 54
BRF, see Biological repopulation 179
Arterial perfusion (AP), 80
factor Complicated hepatic arterial access,
Arterial phase, 11–12
BSA, see Body surface area 56–57
Automated segmentation tools, 101
Burial, 158–159 Cone-beam computed tomography
Autopsy, posttreatment events,
(CBCT), 97
158–159
Constrained constructive
Average absorbed dose to tumor C
optimization (CCO), 207
(Davg), 317–318
Calibrated phantoms, Continuity of care, 269
Average dose endpoint (Davg), 98
quantitative imaging, Contrast-enhanced CT (CECT), 283
244–245 Conventional transcatheter arterial
B
C-arm cone beam CT (CBCT), chemoembolization
BED, see Biological effective dose 13–14, 81 (cTACE), 38
Biliary effects; see also Radiation CBCT, see C-arm cone beam CT CRC, see Colorectal cancer
cholecystits CCO, see Constrained constructive CRChot, see Hot contrast recovery
biliary stricture, 294 optimization coefficient
biloma, 292–293 CECT, see Contrast-enhanced CT CRC metastasis, see Colorectal
cholangitis and transient Celiac artery stenosis, 56–57 carcinoma metastasis
hyperbilirubinemia, Cell survival curve, 171 Cremation, 158–159
293–294 CHA, see Common hepatic artery Cryoablation, 37–38

323
324 Index

cTACE, see Conventional DPK convolution, see Dose-point External beam radiotherapy
transcatheter arterial kernel convolution (EBRT), 41–42, 47, 168,
chemoembolization Drug-eluting bead TACE 285
CTCAE, see Common Terminology (DEB-TACE), 38, 43, 47 Lyman normal tissue
Criteria for Adverse Events DSA, see Digital subtraction complication probability,
Cytotoxic chemotherapy, 34 angiography 183–185
DSM, see Degradable starch principles, 7–8
microspheres tolerance doses from, 182–183
D
DVH, see Dose–volume histogram Extrahepatic radioembolization, 318
D70 dose, 317–318 DW-MRI, see Diffusion-weighted
Davg, see Average absorbed dose to MRI
F
tumor
DEB-TACE, see Drug-eluting bead Falciform artery, 296
E
TACE FDA, see Food and Drug
Degradable starch microspheres EASL, see European Association for Administration
(DSM), 315 the Study of the Liver Femoral artery access, 54
2-Deoxy-2-18fluoroglucose PET/ Eastern Cooperative Oncology Fibrotic remodeling, 287
CT, 283 Group (ECOG) scale, 34 FLR hypertrophy, see Future liver
Diffuse/focal vascular disease, 56 ECOG scale, see Eastern remnant hypertrophy
Diffusion-weighted MRI Cooperative Oncology Folinic acid, fluorouracil,
(DW-MRI), 283 Group scale oxaliplatin (FOLFOX)
Digital subtraction angiography Effective dose, 88–89 chemotherapy, 44
(DSA), 13 Embryological middle hepatic Follow-up imaging assessment
DNA damage, repair process of, artery (eMHA), 12–13 criteria in predicting
168–169 Embryological right hepatic artery treatment response,
Dosage, 88–89 (eRHA), 12–13 287–290
Dosage cart preparation, 148–151 eMHA, see Embryological middle investigational follow-up
Dosage delivery hepatic artery methods, 290–291
dosage cart preparation, Empiric model, for resin role of 18Fdg-PET/CT, 290
148–151 microspheres, 97–98, Food and Drug Administration
exposure rate measurement of 104–105 (FDA), 5, 67, 136
dosage vials, 148 End-hole microcatheter, 18 4Rs of radiation biology, 170–171
SIR-Spheres dosage dispensing EQ, see Equivalent dose Fractional uptake (FU), 99
and preparation, 144–145 Equivalent dose (EQ), 174 FSU, see Functional subunits
TheraSphere dosage dispensing Equivalent uniform biological FU, see Fractional uptake
and preparation, 145–148 effective dose (EUBED), Functional subunits (FSU), 180
Dose calibrator, 142 182 Functional tumor response
Dose deposition around A b source, Equivalent uniform dose (EUD), 182 assessment, 22–23
203–204 eRHA, see Embryological right Future liver remnant (FLR)
Dose heterogeneity, 92 hepatic artery hypertrophy, 117–119,
Dose metrics of interest, 317–318 EUBED, see Equivalent uniform 124–126
Dose-point kernel (DPK) biological effective dose
convolution, 253–254 EUD, see Equivalent uniform dose
G
and local deposition method, European Association for the Study
259–260 of the Liver (EASL), Gastroduodenal artery (GDA),
Dose–response 120–122, 288 12–13
data, 318 EVBT, see Prophylactic coil embolization, 58
function, 180 endovascular Gastrointestinal nontarget
relationship, 19 brachytherapy embolization, 295
Dose-toxicity model, 181 Extended cardiac-torso (XCAT) Gastrointestinal stromal tumors
Dose–volume histogram (DVH), 232 phantom simulation, 224 (GIST), 288
Index 325

Gastrointestinal ulceration, 60 Hepatopulmonary shunting, 59–60 Intralobule dosimetry, in translation


Geometric template, for survey Holmium-166 microspheres, 10–11, invariant trapping
meter measurement, 148 82 from Monte Carlo simulations,
GIST, see Gastrointestinal stromal Hormonal therapy, 34–35 204–205
tumors Hot contrast recovery coefficient from Russell’s law, 205–206
Glass microspheres (TheraSphere), (CRChot), 238 Intraprocedural imaging, 13–15
9, 66, 308 Human serum albumin (HSA), 67, Intratumoral dose heterogeneity, 92
treatment planning, 98–99, 105 310 Ionizing radiation, 169
Glidesheath, 54 Hypertrophy, 193–194 IR, see Interventional radiology
IRE, see Irreversible electroporation
Irreversible electroporation (IRE),
H I
38
HCC, see Hepatocellular carcinoma IAEA Basic Safety Standards, see
Hepatic abscess, 295 International Atomic
K
Hepatic arterial anatomy, 12, 54–56 Energy Agency (IAEA)
Hepatic arterial tree modeling, Basic Safety Standards Killed lobule fraction (KF), 212
207–208 ICC, see Intrahepatic
Hepatic atrophy, 286 cholangiocarcinoma
L
Hepatic malignancy, 34 IEC, see International
Hepatic radioembolization, 3–8 Electrotechnical LDM, see Local deposition method
catheter types and particle-fluid Commission Left gastric artery (LGA), 12–13, 54
dynamics, 18 Imaging of complications Left hepatic artery (LHA), 54
dose–response relationship, 19 biliary effects, 292 LGA, see Left gastric artery
imaging of therapeutic hepatic abscess, 295 Linear quadratic model, 171–173,
microsphere distribution, hepatic toxicity, 291–292 185–189
18–19 Imaging protocol, 69–71 Linear regression analysis of
laboratory toxicity, 19–20 Imaging surveillance protocols, exposure rate, 148
medication and periprocedural 282–283 Liver
care, 17 Immediate postadministration, CT/MRI, 11–12
postembolization syndrome, 21 radiation safety treatment functional anatomy of, 12
pretreatment workup, 11–17 in IR suite, 154–155 hypertrophy, 193
treatment, 17–19 Individual hepatic arterial anatomy, metastasis, 282
treatment-related laboratory and 12–13 radioembolization, scales in,
clinical toxicity, 19–21 Inert particle embolization, 38 200–201
treatment technique, 17–18 Internal pair production of 90Y, resection, 158
tumor response assessment, 230–231 transplantation, 35–36, 43, 46
21–23 International Atomic Energy Liver and tumor volumes, 100
types of microspheres and Agency (IAEA) Basic automatic and semiautomatic
radionuclides, 8–11 Safety Standards, 233 segmentation, 101
with yttrium-90 (90Y) International Electrotechnical manual segmentation, 101–102
microspheres, 281 Commission (IEC), 224 Liver cancer
Hepatic structure, blood outflow in, Interventional radiology (IR), 135 colorectal cancer, 44–45
200–201 Intra-arterial access, 13 hepatic malignancy, 34
Hepatic toxicity, 211–212, 291–292 Intra-arterial infusion, of hepatocellular carcinoma,
dose threshold for, 94 vasoconstrictor 42–44
model, application of, 212–215 angiotensin II, 312 locoregional therapies, see
Hepatocellular carcinoma (HCC), Intrahepatic arterial distribution, Locoregional therapies
4, 42–44, 67, 70, 282 56 neuroendocrine tumor, 45–47
liver transplantation, 35–36 Intrahepatic biliary tree, 292 signs and symptoms, 33
particle embolization, 38 Intrahepatic cholangiocarcinoma systemic therapies, 34–35
Hepatocyte proliferation, 115 (ICC), 4, 47 transarterial therapy, 38–42
326 Index

LKB model, see Lyman–Kutcher– MC methods, see Monte Carlo Necrotic neuroendocrine tumor
Burman model methods (NET) metastases, 284
Lobar therapy, 103–104 mCRC, see Metastatic colorectal NEMA, see National Electrical
Lobectomy dosimetry, 117 cancer Manufacturers
Lobules, 200 Mean absorbed dose Association
Local deposition method (LDM), in hepatic structures, 205 NET, see Neuroendocrine tumor
253, 255–256, 276 tissue, 277 NET metastases, see Necrotic
and alternative radionuclides, Mean lung dose (MLD), 70–72 neuroendocrine tumor
258–259 Median arcuate ligament metastases
dose-point kernel convolution syndrome, 56–57 Neuroendocrine tumor (NET), 4,
and, 259–260 Median time to progression (TTP), 45–47
validation of, 256–258 287 NIST, U.S., see National Institutes
Locoregional therapies (LRT), 282 Medical internal radiation dose of Standards and
cryoablation, 37–38 (MIRD), 71, 99, 221, 277 Technology, U.S.
irreversible electroporation, 38 Medium-energy (ME) collimator, Non–dose-reducing techniques,
liver transplantation, 35–36 223 60
microwave and radiofrequency MELD, see Model for end-stage Nononcologic extrahepatic use, of
ablation, 37 liver disease radioembolization, 318
resection surgery, 35 Metastatic colorectal cancer Nontarget embolization, 296
Low-volume perihepatic ascites, 287 (mCRC), 44–45, 95 Nontarget embolization (NTE),
LSF, see Lung shunt fraction Microdosimetry, 89–90 282, 296
Lung dosimetry, 71–73 Microspheres, 89–91 Nontarget radioembolization,
Lung shunt fraction (LSF), 69, 70, 283 biodistribution studies in liver, techniques to prevent,
Lyman–Kutcher–Burman (LKB) 206–207 58–59
model, 176–180 distribution, 209–211, 211–212 Nonuniform irradiation, 191–193
Lyman model, 176–180 glass, 9 Normal liver endpoints, 94–95
Lyman normal tissue complication holmium-166, 10–11 Normal response to therapy
probability, 183–185 resin, 9–10 radiation lobectomy and
transport modeling, 208–209 segmentectomy, 286
yttrium-90, 8–9 reduction in tumor size and
M
Microwave (MW) ablation, 37 changes in liver volume
TC Macroaggregated albumin
99m MIRD, see Medical internal (beyond 3 months),
(99mTC-MAA), 221–222, radiation dose 285–286
283, 309–310 MLD, see Mean lung dose rim enhancement, necrosis,
flow dynamics and particle size Model for end-stage liver disease and pseudoprogression
effects, 77–78 (MELD), 34 (0–3 months), 284–285
imaging, 66–67 Modified RECIST (mRECIST), 288 sequela of portal hypertension
imaging protocol, 69–71 Monte Carlo (MC) methods, after therapy, 287
lung dosimetry, 71–73 201–203, 252 Normal tissue complication
lung shunt fraction evaluation, intralobule dosimetry from, probability (NTCP)
76–77 204–205 predictions
macroaggregated albumin mRECIST, see Modified RECIST architecture model, 180–181
alternatives, 80–82 clinical cases, with nonuniform
nontarget embolization and irradiation, 191–193
N
radiation penumonitis, hypothesis, of uniform
73–76 National Electrical Manufacturers irradiation, 190–191
physical and biological Association (NEMA) Lyman model, 176–180
properties, 67–69 NU 2-2007 procedure, 235–237 NTCP predictions, see Normal
quantification trends, 69 National Institutes of Standards tissue complication
vs. absorbed dose, 78–80 and Technology (NIST), probability predictions
Manual segmentation, 101–102 U.S., 142–143 NTE, see Nontarget embolization
Index 327

O Postprocedural imaging, in medical Pretreatment workup, for hepatic


management radioembolization, 11–12
Oxygen enhancement ratio (OER),
of complications, 291–296 Progression of disease, 287
169
follow-up imaging assessment Proper hepatic artery (PHA), 12–13,
criteria in predicting 54
treatment response, Prophylactic endovascular
P
287–291 brachytherapy (EVBT),
Parallel architecture model, imaging surveillance protocols, 309
180–181 282–283 Pseudoprogression, 285
Parasitization, 97 normal response to therapy, PSF, see Point-spread function
Parasitized arterial perfusion, 284–287 PTA, see Percutaneous
57–58 posttreatment 90Y imaging, transluminal angioplasty
Particle embolization, 38, 39 296–298 PVE, see Portal vein embolization
Particle-fluid dynamics, 18 with prognostic significance,
Partition model, 99, 139–140 283–284
resin microspheres using, treatment modification with
Q
90Y PET/CT, 299
105–106 Quantitative postradioembolization
PERCIST criteria, see PET Postradioembolization imaging imaging technique
Response Criteria in Solid using diagnostic reporting accuracy of, 231, 245
Tumors criteria 90Y PET/CT, see Yttrium-90 activity measurements of 90Y
Percutaneous transluminal positron emission chloride, 233–234
angioplasty (PTA), 309 tomography with activity measurements of 90Y
PET radiotracer, see Positron integrated CT microspheres, 234–235
emission tomography (90Y PET/CT) with calibrated phantoms,
radiotracer absorbed doses verification, 244–245
PET Response Criteria in Solid 275–277 clinical examples of, 245, 246
Tumors (PERCIST) bremsstrahlung SPECT/CT, decay scheme of 90Y, 230–231
criteria, 290 264–266 PET specifications, see Positron
PHA, see Proper hepatic artery continuity of care, 269 emission tomography
Planar scintigraphy, 70 nontarget absorbed dose, (PET), performance
Point-spread function (PSF), 257, quantification of, 274–275 characteristics
258 nontarget activity vs. noise, phantom study analysis,
Portal vein embolization (PVE), 270–273 231–233
35, 115 target activity, 269–272 Quantitative postradioembolization
Positron emission tomography technical failure, 272–274 imaging, use of, 316–317
(PET), performance technical success, 263 QUEST phantom study, 232,
characteristics Postradioembolization quantitative 238–240, 244
absolute activity calibration, 237 imaging, tumor
degrading factors, 241–243 contouring on, 317
r
image reconstruction, 243–244 Posttreatment 90Y imaging
recovery coefficients, 238–241 evaluating nontarget Radiation biology, 115–116
sensitivity analysis, 235–237 embolization with, Radiation biology, of
spatial resolution, 237–238 296–297 radioembolization,
2D and 3D acquisition mode, 236 prognostication with 90Y PET/ 167–168
Positron emission tomography CT, 297–298 4Rs, 170–171
(PET) radiotracer, 310 Posttreatment patient management radiobiological models, see
Postembolization syndrome, 21, 60 follow-up evaluation, 124 Radiobiological models
Postprocedural 90Y single-photon patient care in, 123–124 repair process, of DNA damage,
emission computed Preparatory angiography, 13–15 168–169
tomography (SPECT/ Pretreatment activity calculations, repopulation, 169
CT), 295 16–17 Radiation cholangitis, 293
328 Index

Radiation cholecystits, 294 Radiation synovectomy, 308–309 Renal artery radioembolization


Radiation-induced damage, Radioactive waste (RARE), 318
171–172 example procedure Renal cell carcinoma (RCC), 318
Radiation-induced liver disease measurements and Reoxygenation, of hypoxic tumor
(RILD), 60, 94, 291 calculations, 160–162 areas, 169–170
Radiation lobectomy, 286 residual activity assay, 159–160 Resectability, 193
definition, 114–115 storage for decay and disposal, Resection surgery, 35, 43
dosimetry, 117 162–163 Residual activity assay, 159–160
and future liver remnant Radiobiological models, 181–182 Resin microspheres (SIR-Spheres),
hypertrophy, 117–119, biological effective dose, 9–10, 66, 308
124–126 173–174 Response evaluation criteria in
overview, 113–114 external beam radiotherapy, solid tumors (RECIST),
patient selection, 116 see External beam 284, 287–288
radiation biology and, 115–116 radiotherapy RFA, see Radiofrequency ablation
treatment planning and linear quadratic model, see RGA, see Right gastric artery
delivery, 114 Linear quadratic model RHA, see Right hepatic artery
Radiation pneumonitis (RP), 60, normal tissue complication RHA, see Right hepatic artery
75–76, 296 probability predictions, Right gastric artery (RGA), 54–55,
Radiation release by patients see Normal tissue 59, 295
hospital admission, 157 complication probability Right hepatic artery (RHA), 54–56
to public, 156–157 predictions RILD, see Radiation-induced liver
Radiation safety concerns tumor control probability, disease
dosage delivery, 142–151 174–176 RP, see Radiation pneumonitis
dosage in interventional various models compared, Russell’s law, in translation
radiology (IR) suite, 189–190 invariant trapping,
152–155 Radioembolization, 38–40 205–206
posttreatment events, 158–159 colorectal cancer, 44–45
radioactive waste, 159–163 hepatocellular carcinoma, 43
S
release of radioactive patient, intrahepatic
155–157 cholangiocarcinoma, 47 SBRT, see Stereotactic body
written directive for toxicities and complications, radiation therapy
administration byproduct 122–123 Scout dose distribution, imaging of,
material, 136–142 Radioembolization-induced liver 15–16
Radiation safety treatment, in disease (REILD), 40, 123, Segmental therapy, 106–109
interventional radiology 291–292 Segmental vascularization, 13
(IR) suite Radioembolization prescription Segmentectomy, 286
administration, assembly of methods, 140–14 dosimetry, 120
infusion system, 153–154 Radiofrequency ablation (RFA), 37 Selective internal radiation therapy
immediate postadministration, Radiolabeled microspheres, 142 (SIRT), 7
154–155 Radionuclides, 8–11 Semiautomatic segmentation, 101
immediate preadministration, RARE, see Renal artery Sequela of portal hypertension, 287
151–153 radioembolization SF, see Surviving fraction
Radiation segmentectomy, 4, 286 RCC, see Renal cell carcinoma SIMIND, see Simulations of
definition, 119–120 RCs, see Recovery coefficients imaging nuclear detectors
outcome data, 120–122 RECIST, see response evaluation Simulations of imaging nuclear
overview, 113–114 criteria in solid tumors detectors (SIMIND), 224
patient selection, 120 Recovery coefficients (RCs), SIR-Spheres
sample case, 126–128 238–241 dosage administration, 153–154
toxicities, 123 Redistribution of cells, 170 infusion system, 148
treatment planning and REILD, see Radioembolization- prescription, 138–140
delivery, 114 induced liver disease strontium-90 impurity in, 163
Index 329

SIRT, see Selective internal Monte Carlo method, 252 U


radiation therapy voxel S-value Medical Internal
U.S. Food and Drug Administration
SMA, see Superior mesenteric Radiation Dose, 254–255
(USFDA), 116
artery Thrombosis, risk of, 54
90Y SPECT/CT imaging, 222, 225, 226 Tissue damage, 171
Standard arterial anatomy, of adult T:N, see Tumor-to-normal liver ratio V
liver, 12 Total EDE (TEDE), 156–157
Standard end-hole microcatheter, 13 Toxicity evaluation, 195 Variable infusion techniques, 286
Stasis, 154 Transarterial brachytherapy, 282 Vascular considerations
Stereotactic body radiation therapy Transarterial chemoembolization angiogenesis, 61
(SBRT), 8, 42 (TACE), 38, 47, 60, 115 arterial access, 54
Superior mesenteric artery (SMA), Transarterial locoregional complicated hepatic arterial
12–13 therapies, 282 access, 56–57
Superior mesenteric artery Transarterial therapy, 38–42, complications, 60–61
technique, 57 46–47 hepatic arterial anatomy,
Surgical therapy Transient hyperbilirubinemia, 54–56
colorectal cancer, 44 293–294 parasitized arterial perfusion,
neuroendocrine tumor, 45–46 Transplant, 158–159 57–58
Surviving fraction (SF) of cells, Treatment planning, in 90Y techniques to minimize
172, 173 radioembolization, hepatopulmonary
Systemic therapy 309–311 shunting, 59–60
cytotoxic chemotherapy, 34 Treatment planning models, techniques to prevent nontarget
hepatocellular carcinoma, 99–100 radioembolization,
43–44 TTP, see Median time to 58–59
hormonal therapy, 34–35 progression Vascular endothelial growth factor
neuroendocrine tumor, 46 Tumor ablation (VEGF), 61
targeted therapy, 35 colorectal cancer, 45 VEGF, see Vascular endothelial
cryoablation, 37–38 growth factor
hepatocellular carcinoma, 43 Volume of interest (VOI) activity,
t 224
intrahepatic
TACE, see Transarterial cholangiocarcinoma, 47 Voxel-based absorbed dose, 204
chemoembolization irreversible electroporation, 38 Voxel S-value (VSV) medical
Targeted therapy, 35 microwave and radiofrequency internal radiation dose
TCP, see Tumor control probability ablation, 37 (MIRD) approach,
TEDE, see Total EDE neuroendocrine tumor, 46 254–255
Therapeutic microsphere Tumor angiogenesis, 312
distribution, imaging of, Tumor control, 176
W
18–19 process of, 171
TheraSphere Tumor control probability (TCP), World Health Organization (WHO)
day-of-therapy, example of, 161 174–176 criteria, 284, 287–288
dosage, administration of, 154 Tumoricidal endpoints, 95 Worsened portal hypertension,
infusion system, 148–149 Tumor response assessment, consequences of, 287
preparation of, 147 22–23 Worsening thrombocytopenia,
prescription/preparation, 140 Tumor response, to therapy, 282 287
Thick rim enhancement, 284–285 Tumor targeting, methods to Written directive, administration
Three-dimensional (3D) image- enhance, 314–315 of byproduct material
based dosimetry Tumor-to-normal liver ratio (T:N), authorized user, use of
dose-point kernel convolution, 95–97, 277 radioactive material,
253–254 pharmaceutical methods, 137–138
local deposition method, see Local 312–313 radioembolization prescription
deposition method (LDM) physical techniques, 313–314 methods, 140–141
330 Index

reportable medical event, Y 90Y bremsstrahlung imaging


141–142 planar, 222–223
Yttrium-90 positron emission
SIR-Spheres prescription, SPECT and SPECT/CT,
tomography with
138–140 223–225
integrated CT
TheraSphere prescription, 140 90Y chloride (90YCl ), activity
3
(90Y PET/CT)
yttrium-90 microspheres measurements, 233–234
bremsstrahlung SPECT/CT,
absorbed dose-related 90Y microspheres, 137–138, 282
264–265, 267
properties, 138 99mTC-macroaggregated
economics of, 277–278
albumin vs., 76–80
prognostication with,
activity measurements,
297–298
X 234–235
technical challenges of, 265,
XCAT phantom simulation, see 267–268
Extended cardiac-torso treatment modification with,
phantom simulation 299

You might also like